1
|
Liang Y, Zhang Q, Qian JR, Li SS, Liu QF. Inflammation-Induced Klotho Deficiency: A Possible Key Driver of Chronic Kidney Disease Progression. Int J Gen Med 2025; 18:2507-2520. [PMID: 40376197 PMCID: PMC12080484 DOI: 10.2147/ijgm.s513497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 05/05/2025] [Indexed: 05/18/2025] Open
Abstract
Chronic kidney disease (CKD) is influenced by inflammation, a critical factor in its progression. However, the underlying mechanism through which inflammation contributes to CKD is still obscure. The Klotho protein, which is predominantly found in the kidneys, is known for its protective functions, including anti-inflammatory, anti-aging, antioxidant, and anti-fibrotic effects. A myriad of studies have suggested that inflammation in CKD leads to a decrease in Klotho expression, diminishing Klotho protection capabilities and exacerbating kidney damage, thereby promoting CKD progression. These findings suggest that Klotho deficiency could be a crucial link between inflammation and CKD progression. However, the mechanism regarding their relationship is still unclear. The reduction in Klotho due to inflammation may be attributed to epigenetic mechanisms, such as DNA methylation, histone deacetylation, transcription factor, microRNA (miRNA) regulation and long non-coding RNA (lncRNA) regulation or non-epigenetic factors, such as endoplasmic reticulum (ER) stress and ER-associated degradation (ERAD), which affect Klotho protein metabolism. Through these pathways, inflammation triggers a decrease in Klotho expression, further driving CKD progression. Notably, Klotho also exerts a strong anti-inflammatory effect by inhibiting key inflammatory factors and pathways, suggesting that there is intricate crosstalk between inflammatory factors and Klotho in CKD. This review highlights how inflammation suppresses the expression of Klotho and further contributes to the development and exacerbation of CKD. By focusing on the interplay between inflammation and Klotho, the present review provides novel potential therapeutic strategies such as correcting epigenetic and non-epigenetic abnormalities for treating CKD by targeting this specific axis.
Collapse
Affiliation(s)
- Yan Liang
- Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, Jiangsu, 215300, People’s Republic of China
| | - Qi Zhang
- Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, Jiangsu, 215300, People’s Republic of China
| | - Jing-Rong Qian
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, People’s Republic of China
| | - Sha-Sha Li
- Clinical Research and Laboratory Centre, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, People’s Republic of China
| | - Qi-Feng Liu
- Gusu School, Nanjing Medical University, The First People’s Hospital of Kunshan, Kunshan, Jiangsu, 215300, People’s Republic of China
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, People’s Republic of China
| |
Collapse
|
2
|
Wang C, Zhou J, Jia P, Yang Y, Song R, Zheng X, Zhang H, Li Y. Joint proteomic and metabolomic analysis reveals renal metabolic remodeling of chronic heart failure mice. J Pharm Biomed Anal 2025; 255:116641. [PMID: 39731929 DOI: 10.1016/j.jpba.2024.116641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/01/2024] [Accepted: 12/15/2024] [Indexed: 12/30/2024]
Abstract
Pharmacologic intervention in chronic heart failure (HF) with renal insufficiency is one of the clinical challenges due to the fact that the mechanisms of cardio-renal interactions in chronic heart failure (CHF) progressing have not been fully revealed. In this paper, C57BL/6 mice were applied thoracic aortic narrowing surgery to establish pressure overload CHF model. Cardiac function, serum markers, renal pathologic changes and kidney metabolism were analyzed at 4th, 8th, 12th, and 16th week after surgery respectively to evaluate the heart-Kidney pathologic overlap. Kidney proteomic analysis was performed at 16th week after operation. As a result, renal hypofiltration and exacerbation of pathological damage was observed accompanying cardiac function deterioration after 12th week. 66 differentially expressed proteins and 13 differential metabolites were found to be involved in the cardio-renal pathological overlap. Joint proteomic and metabolomic analysis revealed that signal pathways like Phosphatidylinositol signaling system, Glucagon signaling pathway, the Glyoxylate and dicarboxylate metabolism; DEPs of Pten, Mtmr4, PLC and CPT1, differential metabolites like aspartic acid and isocitrate deserve further investigation.
Collapse
Affiliation(s)
- Chunliu Wang
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China; Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China
| | - Jie Zhou
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Pu Jia
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, Shaanxi, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, Shaanxi, China
| | - Ruixue Song
- Xi'an Research Institute of Chinese Lacquer, Xi'an, Shaanxi, China
| | - Xiaohui Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, Shaanxi, China
| | - Hong Zhang
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China; Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China.
| | - Ye Li
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China; Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China.
| |
Collapse
|
3
|
Bala N, Rafay RH, Glover SC, Alli AA. Activity of Various Cathepsin Proteases and Enrichment of Klotho Protein in the Urine and Urinary Extracellular Vesicles After SARS-CoV-2 Infection. Viruses 2024; 17:25. [PMID: 39861814 PMCID: PMC11768607 DOI: 10.3390/v17010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/20/2024] [Accepted: 12/25/2024] [Indexed: 01/27/2025] Open
Abstract
Background: The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is responsible for causing the Coronavirus disease 2019 (COVID-19) outbreak. While mutations cause the emergence of new variants, the ancestral SARS-CoV-2 strain is unique among other strains. Methods: Various clinical parameters, the activity of cathepsin proteases, and the concentration of various proteins were measured in urine samples from COVID-19-negative participants and COVID-19-positive participants. Urinary extracellular vesicles (uEVs) were isolated from urine samples from the two groups and used for proteomic analysis and subsequent pathway analyses. Results: Activity levels of cathepsin S and L were greater in the urine of COVID-19-positive participants. The concentration of C-reactive protein, transmembrane serine protease 2, and klotho protein were significantly greater in the urine of COVID-19-positive participants. There was a greater amount of uEVs in the COVID-19 group and klotho protein was found to be enriched in uEVs from the COVID-19 group. Pathway analyses of the proteomics data showed most of the identified proteins were involved in signal transduction, stress response, protein metabolism, and transport. The identified proteins were predominantly associated with cellular membranes and with function of the cytoskeleton, enzyme regulation, and signal transduction. Conclusions: Taken together, our data identify novel urinary biomarkers that could be used to further investigate the long-term effects of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Niharika Bala
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida College of Medicine, Gainesville, FL 32608, USA; (N.B.); (R.H.R.)
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32608, USA
| | - Ramish H. Rafay
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida College of Medicine, Gainesville, FL 32608, USA; (N.B.); (R.H.R.)
| | - Sarah C. Glover
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA;
| | - Abdel A. Alli
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida College of Medicine, Gainesville, FL 32608, USA; (N.B.); (R.H.R.)
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32608, USA
| |
Collapse
|
4
|
Behzadi P, Cuevas RA, Crane A, Wendling AA, Chu CC, Moorhead WJ, Wong R, Brown M, Tamakloe J, Suresh S, Salehi P, Jaffe IZ, Kuipers AL, Lukashova L, Verdelis K, St Hilaire C. Rapamycin Reduces Arterial Mineral Density and Promotes Beneficial Vascular Remodeling in a Murine Model of Severe Medial Arterial Calcification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.01.606196. [PMID: 39149364 PMCID: PMC11326142 DOI: 10.1101/2024.08.01.606196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Peripheral artery disease (PAD) is the narrowing of the arteries that carry blood to the lower extremities. PAD has been traditionally associated with atherosclerosis. However, recent studies have found that medial arterial calcification (MAC) is the primary cause of chronic limb ischemia below the knee. MAC involves calcification of the elastic fibers surrounding smooth muscle cells (SMCs) in arteries. Matrix GLA protein (MGP) inhibits vascular calcification by binding circulating calcium and preventing hydroxyapatite crystal deposition, while also modulating osteogenic signaling by blocking BMP-2 activation of RUNX2. Mgp -/- mice develop severe MAC and die around 8 weeks after birth due to aortic rupture or heart failure. We previously discovered a rare genetic disease Arterial Calcification due to Deficiency in CD73 (ACDC) in which patients present with extensive MAC in their lower extremity arteries. Using a patient-specific induced pluripotent stem cell model we found that rapamycin inhibited calcification. Here we investigated whether rapamycin could reduce MAC in vivo using the Mgp -/- murine model. Mgp +/+ and Mgp -/- mice received 5mg/kg rapamycin or vehicle. Calcification content was assessed via microCT, and vascular morphology and extracellular matrix content assessed histologically. Immunostaining and western blot analysis were used to examine SMC phenotype and extracellular matrix content. Rapamycin prolonged Mgp -/- mice lifespan, decreased mineral density in the arteries, maintained SMC contractile phenotype, and improved vessel structure, however, calcification volume was unchanged. Mgp -/- mice with SMC-specific deletion of Raptor or Rictor, did not recapitulate treatment with rapamycin. These findings suggest rapamycin promotes beneficial vascular remodeling in vessels with MAC.
Collapse
Affiliation(s)
- Parya Behzadi
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rolando A Cuevas
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alex Crane
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andrew A Wendling
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Claire C Chu
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - William J Moorhead
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ryan Wong
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mark Brown
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Joshua Tamakloe
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Swathi Suresh
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Payam Salehi
- CardioVascular Center, Vascular Surgery, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111-1800, USA
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111-1800, USA
| | - Allison L Kuipers
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lyudmila Lukashova
- Departments of Endodontics and Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Konstantinos Verdelis
- Departments of Endodontics and Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cynthia St Hilaire
- Department of Medicine, Division of Cardiology, and the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
5
|
Ranjbar N, Ebrahimi Behnam B, Mesgari Abbasi M, Esmaeili M, Jolfaei F, Mohammadian J, Rashtchizadeh N, Ghorbanihaghjo A, Raeisi S. The possible antioxidative effects of ketogenic diet by modifying brain klotho expression: a rat model study. Nutr Neurosci 2024:1-7. [PMID: 39674922 DOI: 10.1080/1028415x.2024.2436817] [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: 12/17/2024]
Abstract
Objectives: The ketogenic diet (KD) has long been used as an alternative nonpharmacological therapy to manage pharmacoresistant epilepsy. The anticonvulsant mechanisms of KD have yet to be fully elucidated. The present study explored whether a KD could exert antioxidative effects by altering brain Klotho (Kl) gene expression.Methods: Thirty male rats were divided into three groups: the normal diet (ND) group received standard rat chow; the calorie-restricted diet (CRD) group was maintained at 90% of the calculated energy need; and the KD group received a diet composed of 8% protein, 2% carbohydrates, and 90% fat (per calorie macronutrient). The levels of β-hydroxybutyrate (BHB) in the serum, Kl gene expression in the brain, and Kl protein, malondialdehyde (MDA), and protein carbonyl (PC) levels in the serum and brain were evaluated by standard methods.Results: The serum BHB levels in the KD group were significantly greater than those in the ND and CRD groups (p < 0.001). The Kl expression in the brain was significantly greater in the KD group than in the ND group (p = 0.028). The brain MDA levels in the KD group were significantly lower than those in the ND group (p = 0.006). Elevated BHB was positively correlated with brain Kl expression (r = 0.668, p < 0.001). The brain MDA levels were negatively correlated with brain Kl expression (r = -0.531, p = 0.003) and serum BHB levels (r = 0.472, p = 0.020).Discussion: KD might exert antioxidative effects by increasing BHB and upregulating Kl in the brain. This could be considered a possible anticonvulsant mechanism of KD.
Collapse
Affiliation(s)
- Nasrin Ranjbar
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahador Ebrahimi Behnam
- Department of Biochemistry, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | | | - Mahsa Esmaeili
- Department of Biological Sciences, Faculty of Basic Sciences, Higher Education Institute of Rab-Rashidi, Tabriz, Iran
| | - Fatemeh Jolfaei
- Department of Biological Sciences, Faculty of Basic Sciences, Higher Education Institute of Rab-Rashidi, Tabriz, Iran
| | - Jamal Mohammadian
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Amir Ghorbanihaghjo
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Raeisi
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Zhang L, Li D, Aierken Y, Zhang J, Liu Z, Lin Z, Jiang L, Li Q, Wu Y, Liu Y. KPV and RAPA Self-Assembled into Carrier-Free Nanodrugs for Vascular Calcification Therapy. Adv Healthc Mater 2024; 13:e2402320. [PMID: 39252648 DOI: 10.1002/adhm.202402320] [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: 06/25/2024] [Revised: 08/14/2024] [Indexed: 09/11/2024]
Abstract
Cardiovascular disease (CVD) is a leading cause of death globally, and vascular calcification (VC) is an important independent risk factor for predicting CVD. Currently, there are no established therapeutic strategies for the treatment of VC. Although recognized combination therapies of nanomedicines can provide effective strategies for disease treatment, the clinical application of nanomedicines is limited because of their complex preparation processes, low drug loading rates, and unpredictable safety risks. Thus, developing a simple, efficient, and safe nanodrug to simultaneously regulate inflammation and autophagy may be a promising strategy for treating VC. Herein, an anti-inflammatory peptide (lysine-proline-valine peptides, KPV) and the autophagy activator rapamycin (RAPA) are self-assembled to form new carrier-free spherical nanoparticles (NPs), which shows good stability and biosafety. In vivo and in vitro, KPV-RAPA NPs significantly inhibit VC in mice compared to the other treatment groups. Mechanistically, KPV-RAPA NPs inhibit inflammatory responses and activated autophagy. Therefore, this study indicates that the new carrier-free KPV-RAPA NPs have great potential as therapeutic agents for VC combination therapy, which can promote the development of nanodrugs for VC.
Collapse
Affiliation(s)
- Li Zhang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Dongze Li
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Yierpani Aierken
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Jie Zhang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Zhenyu Liu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Zipeng Lin
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Longqi Jiang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Qingzhu Li
- Department of General Surgery, Gulin People's Hospital, Luzhou, 646000, China
| | - Ya Wu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Yong Liu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| |
Collapse
|
7
|
Hung Vu M, Shiwakoti S, Ko JY, Bang G, Lee E, Kim E, Park SH, Park EH, Woo Kim C, Young Kim J, Sim HH, Chang K, Kim MS, Oak MH. Niclosamide attenuates calcification in human heart valvular interstitial cells through inhibition of the AMPK/mTOR signaling pathway. Biochem Pharmacol 2024; 230:116614. [PMID: 39515588 DOI: 10.1016/j.bcp.2024.116614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 10/01/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Calcific aortic valve disease (CAVD) is a considerable health burden with a lack of effective therapeutic options. There is an urgent need to develop interventions that inhibit the osteogenic transformation of valvular interstitial cells (VICs) and delay the calcification process. Niclosamide, an FDA-approved anti-helminthic drug, has emerged as a promising candidate that demonstrates a negative regulatory effect on porcine VICs calcification. However, its molecular mechanism in human VICs (hVICs) remains to be investigated. In this study, high-resolution mass spectrometry-based proteomics and phosphoproteomics were employed, and 8373 proteins and 3697 phosphosites were identified in hVICs treated with a pro-calcifying medium and niclosamide. The quantitative proteomic and phosphoproteomic analysis resulted in the identification of calcification markers and osteogenesis-associated proteins. Bioinformatic analysis of the protein-protein interaction network and affected kinase prediction revealed that the AMPK/mTOR/p70S6K signaling cascade was altered upon calcific induction and niclosamide treatment. Further validation indicated that niclosamide inhibited the calcification of hVICs by targeting the mammalian target of the rapamycin (mTOR) signaling pathway. This study provides the first evidence that niclosamide could prevent osteoblastic differentiation in hVICs partially through the inhibition of the AMPK/mTOR/p70S6k signaling pathway, thereby mitigating hVICs calcification. These findings present a foundation for potential therapeutic strategies to impede the progression of CAVD and provide valuable insights into the pharmacological effects of niclosamide on human VICs.
Collapse
Affiliation(s)
- Minh Hung Vu
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Saugat Shiwakoti
- College of Pharmacy, Mokpo National University, 1666 Yeongsan-Ro, Cheonggye-Myeon, Muan-Gun, Jeonnam 58554, Republic of Korea
| | - Ju-Young Ko
- College of Pharmacy, Mokpo National University, 1666 Yeongsan-Ro, Cheonggye-Myeon, Muan-Gun, Jeonnam 58554, Republic of Korea
| | - Geul Bang
- Digital Omics Research Center, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Eunmi Lee
- Cardiovascular Research Institute for Intractable Disease, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Eunmin Kim
- Cardiovascular Research Institute for Intractable Disease, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sin-Hee Park
- Cardiovascular Research Institute for Intractable Disease, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Eun-Hye Park
- Cardiovascular Research Institute for Intractable Disease, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Chan Woo Kim
- Cardiovascular Research Institute for Intractable Disease, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin Young Kim
- Digital Omics Research Center, Korea Basic Science Institute, Ochang, 28119, Republic of Korea; Critical Diseases Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Hwan-Hee Sim
- College of Pharmacy, Mokpo National University, 1666 Yeongsan-Ro, Cheonggye-Myeon, Muan-Gun, Jeonnam 58554, Republic of Korea
| | - Kiyuk Chang
- Cardiovascular Research Institute for Intractable Disease, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; Division of Cardiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Min-Sik Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea.
| | - Min-Ho Oak
- College of Pharmacy, Mokpo National University, 1666 Yeongsan-Ro, Cheonggye-Myeon, Muan-Gun, Jeonnam 58554, Republic of Korea.
| |
Collapse
|
8
|
Li Q, Wang P, Gong Y, Xu M, Wang M, Luan R, Liu J, Li X, Shao Y. α-Klotho prevents diabetic retinopathy by reversing the senescence of macrophages. Cell Commun Signal 2024; 22:449. [PMID: 39327553 PMCID: PMC11426092 DOI: 10.1186/s12964-024-01838-w] [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: 07/09/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Diabetic retinopathy (DR) is a common microvascular complication of diabetes mellitus (DM) and a significant cause of acquired blindness in the working-age population worldwide. Aging is considered as an important risk factor for DR development. Macrophages in aged mice bear typical M2 marker proteins but simultaneously express a pro-inflammatory factor profile. This may explain why the level of intraocular inflammation does not decrease during proliferative diabetic retinopathy (PDR) despite the occurrence of neovascularization and fibrosis (M2 activation). α-Klotho (KL) was originally discovered as a soluble anti-aging factor, which is mainly expressed in kidney tubular epithelium, choroid plexus in the brain and secreted in the blood. However, the role of KL in DR pathophysiology has not been previously reported. METHODS Type 1 (streptozotocin [STZ]-induced) and type 2 (a high-fat diet along with a low dose of STZ) diabetic mouse models were established and injected with or without KL adenovirus via the tail vein for 12 weeks. Vldlr-/- mice were injected intravitreally with or without soluble KL protein from P8 to P15. The retinal structure and function were analyzed by electroretinogram and optical coherence tomography. The neovascular lesions were analyzed by retinal flat mount and RPE flat mount. The senescence markers, macrophage morphology, and KL expression levels were detected by immunofluorescence staining. A cell model was constructed using RAW264.7 cells stimulated by 4-hydroxynonenal (4HNE) and transfected with or without KL adenovirus. The senescence-associated secretory phenotypes were detected by qRT-PCR. Senescence was detected by SA-β-Gal staining. Serum, aqueous humor, and vitreous humor KL levels of proliferative diabetic retinopathy (PDR) patients were measured by enzyme-linked immunosorbent assay. Quantitative proteomics and bioinformatics were applied to predict the change of proteins and biological function after overexpression of KL in macrophages. The effects of KL on the HECTD1 binding to IRS1 were analyzed by bioinformatics, molecular docking, and Western Blot. RESULTS Serum, aqueous humor, and vitreous humor KL levels were lower in patients with PDR than in those with cataracts. KL relieved the retinal structure damage, improved retina function, and inhibited retinal senescence in diabetic mice. KL administration attenuated the neovascular lesions in VLDLR-/- mice by decreasing the secretion of VEGFA and FGF2 from macrophages. KL also protected RAW264.7 cells from 4HNE-induced senescence. Additionally, it inhibited E3 ubiquitin ligase HECTD1 expression in both diabetic mouse peripheral blood mononuclear cells and 4HNE-treated RAW264.7 cells. KL inhibited HECTD1 binding to IRS1 and reduced the ubiquitination of IRS1. CONCLUSIONS Macrophage aging is involved in DM-induced retinopathy. KL alleviates DM-induced retinal macrophage senescence by downregulating HECTD1 and decreasing IRS1 ubiquitination and degradation. Meanwhile, KL administration attenuated the neovascular lesions by altering the activation state of macrophages and decreasing the expression of VEGFA and FGF2.
Collapse
Affiliation(s)
- Qingbo Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
| | - Peiyu Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
| | - Yi Gong
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
| | - Manhong Xu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
| | - Manqiao Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
| | - Rong Luan
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China
| | - Juping Liu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
| | - Yan Shao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute & School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Nankai District, No. 251, Fukang Road, Tianjin, 300384, China.
- University of Tibetan Medicine, Lhasa, 850000, China.
| |
Collapse
|
9
|
Hosseininasab SS, Dhiaa SM, Shahrtash SA, Lak M, Faghihkhorasani A, Mahdi F. The interaction between klotho protein and epigenetic alteration in diabetes and treatment options. J Diabetes Metab Disord 2024; 23:333-341. [PMID: 38932867 PMCID: PMC11196449 DOI: 10.1007/s40200-024-01387-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/11/2024] [Indexed: 06/28/2024]
Abstract
Introduction Klotho is a membrane protein predominantly expressed in the kidneys, and its discovery was serendipitously made through gene-targeting experiments conducted on mice. Klotho has a favorable role in the regulation of multiple cellular processes, such as aging, oxidative stress, inflammation, and apoptosis. This regulation occurs through the targeting of diverse signaling molecules, cell membrane receptors, and ion channels, achieved by physical contacts or enzymatic activities of Klotho. This review examines the role of Klotho in the epigenetic regulation of molecules associated with diabetes. Methods Authors conducted a thorough literature search using the PubMed®, Web of Science™, and Scopus®. Relevant articles up to September 2023, published in the English language were considered. We reviewed research databases searching for studies that included keywords klotho, epigenetic, and diabetes. Results 14 related papers about epigenetic modification of proteins involved in diabetes pathogenesis were selected to be included in this narrative review. In the studies, the kidney was the most investigated organ regarding this correlation. Also, phosphorylation and methylation were the common epigenetic modifications of proteins by Klotho. Conclusion Klotho has a significant role in the maturation of adipocytes and the regulation of systemic glucose metabolism, exhibiting a strong association with the pathogenesis of diabetes. Both epigenetic alterations and the modulation of protein phosphorylation by Klotho play significant roles in the regulation of Klotho expression and the modulation of other molecules implicated in the etiology of diabetes.
Collapse
Affiliation(s)
| | | | | | - Mehrnoosh Lak
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Fatemeh Mahdi
- Department of Internal Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
10
|
Poursistany H, Azar ST, Azar MT, Raeisi S. The current and emerging Klotho-enhancement strategies. Biochem Biophys Res Commun 2024; 693:149357. [PMID: 38091839 DOI: 10.1016/j.bbrc.2023.149357] [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: 09/05/2023] [Revised: 11/24/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024]
Abstract
Klotho is well known as a gene with antiaging properties. It has membrane and soluble forms, providing a unique system that controls various metabolic processes essential to health and disease. Klotho deficiency has been revealed to be associated with various aging-related disorders. Based on its various known and unknown protective properties, upregulating the Klotho gene may be a possible therapeutic and/or preventive approach in aging-related complications. Some agents, such as hormonal compounds, renin-angiotensin system inhibitors, antioxidants, peroxisome proliferator-activated receptor gamma (PPAR-γ) agonists, statins, vitamin D receptor agonists, antioxidants, anti-inflammatory agents, mammalian target of rapamycin (mTOR) signaling inhibitors, and receptor-interacting serine/threonine-protein kinase 1 (RIPK1) inhibitors, can possibly lead to the upregulation and elevation of Klotho levels. Demethylation and deacetylation of the Klotho gene can also be considered other possible Klotho-enhancement methods. Some emerging techniques, such as RNA modifications, gene therapy, gene editing, and exosome therapy, probably have the potential to be applied for increasing Klotho. In the present study, these current and emerging Klotho-enhancement strategies and their underlying mechanisms were comprehensively reviewed, which could highlight some potential avenues for future research.
Collapse
Affiliation(s)
- Haniyeh Poursistany
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Tabibi Azar
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Mahsan Tabibi Azar
- Student Research Committee, Islamic Azad University, Tabriz Branch, Tabriz, Iran
| | - Sina Raeisi
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Wang X, Wang Z, He J. Similarities and Differences of Vascular Calcification in Diabetes and Chronic Kidney Disease. Diabetes Metab Syndr Obes 2024; 17:165-192. [PMID: 38222032 PMCID: PMC10788067 DOI: 10.2147/dmso.s438618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/21/2023] [Indexed: 01/16/2024] Open
Abstract
Presently, the mechanism of occurrence and development of vascular calcification (VC) is not fully understood; a range of evidence suggests a positive association between diabetes mellitus (DM) and VC. Furthermore, the increasing burden of central vascular disease in patients with chronic kidney disease (CKD) may be due, at least in part, to VC. In this review, we will review recent advances in the mechanisms of VC in the context of CKD and diabetes. The study further unveiled that VC is induced through the stimulation of pro-inflammatory factors, which in turn impairs endothelial function and triggers similar mechanisms in both disease contexts. Notably, hyperglycemia was identified as the distinctive mechanism driving calcification in DM. Conversely, in CKD, calcification is facilitated by mechanisms including mineral metabolism imbalance and the presence of uremic toxins. Additionally, we underscore the significance of investigating vascular alterations and newly identified molecular pathways as potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Xiabo Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Jianqiang He
- Department of Nephrology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| |
Collapse
|
12
|
Kang JH, Kawano T, Murata M, Toita R. Vascular calcification and cellular signaling pathways as potential therapeutic targets. Life Sci 2024; 336:122309. [PMID: 38042282 DOI: 10.1016/j.lfs.2023.122309] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/04/2023]
Abstract
Increased vascular calcification (VC) is observed in patients with cardiovascular diseases such as atherosclerosis, diabetes, and chronic kidney disease. VC is divided into three types according to its location: intimal, medial, and valvular. Various cellular signaling pathways are associated with VC, including the Wnt, mitogen-activated protein kinase, phosphatidylinositol-3 kinase/Akt, cyclic nucleotide-dependent protein kinase, protein kinase C, calcium/calmodulin-dependent kinase II, adenosine monophosphate-activated protein kinase/mammalian target of rapamycin, Ras homologous GTPase, apoptosis, Notch, and cytokine signaling pathways. In this review, we discuss the literature concerning the key cellular signaling pathways associated with VC and their role as potential therapeutic targets. Inhibitors to these pathways represent good candidates for use as potential therapeutic agents for the prevention and treatment of VC.
Collapse
Affiliation(s)
- Jeong-Hun Kang
- National Cerebral and Cardiovascular Center Research Institute, 6-1 Shinmachi, Kishibe, Suita, Osaka 564-8565, Japan.
| | - Takahito Kawano
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masaharu Murata
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Riki Toita
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka, 563-8577, Japan; AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, AIST, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
13
|
Edmonston D, Grabner A, Wolf M. FGF23 and klotho at the intersection of kidney and cardiovascular disease. Nat Rev Cardiol 2024; 21:11-24. [PMID: 37443358 DOI: 10.1038/s41569-023-00903-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/13/2023] [Indexed: 07/15/2023]
Abstract
Cardiovascular disease is the leading cause of death in patients with chronic kidney disease (CKD). As CKD progresses, CKD-specific risk factors, such as disordered mineral homeostasis, amplify traditional cardiovascular risk factors. Fibroblast growth factor 23 (FGF23) regulates mineral homeostasis by activating complexes of FGF receptors and transmembrane klotho co-receptors. A soluble form of klotho also acts as a 'portable' FGF23 co-receptor in tissues that do not express klotho. In progressive CKD, rising circulating FGF23 levels in combination with decreasing kidney expression of klotho results in klotho-independent effects of FGF23 on the heart that promote left ventricular hypertrophy, heart failure, atrial fibrillation and death. Emerging data suggest that soluble klotho might mitigate some of these effects via several candidate mechanisms. More research is needed to investigate FGF23 excess and klotho deficiency in specific cardiovascular complications of CKD, but the pathophysiological primacy of FGF23 excess versus klotho deficiency might never be precisely resolved, given the entangled feedback loops that they share. Therefore, randomized trials should prioritize clinical practicality over scientific certainty by targeting disordered mineral homeostasis holistically in an effort to improve cardiovascular outcomes in patients with CKD.
Collapse
Affiliation(s)
- Daniel Edmonston
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Alexander Grabner
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Myles Wolf
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
14
|
Espartero A, Vidal A, Lopez I, Raya AI, Rodriguez M, Aguilera-Tejero E, Pineda C. Rapamycin downregulates α-klotho in the kidneys of female rats with normal and reduced renal function. PLoS One 2023; 18:e0294791. [PMID: 38015969 PMCID: PMC10684065 DOI: 10.1371/journal.pone.0294791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/07/2023] [Indexed: 11/30/2023] Open
Abstract
Both mTOR and α-klotho play a role in the pathophysiology of renal disease, influence mineral metabolism and participate in the aging process. The influence of mTOR inhibition by rapamycin on renal α-klotho expression is unknown. Rats with normal (controls) and reduced (Nx) renal function were treated with rapamycin, 1.3 mg/kg/day, for 22 days. The experiments were conducted with rats fed 0.6% P diet (NP) and 0.2% P diet (LP). Treatment with rapamycin promoted phosphaturia in control and Nx rats fed NP and LP. A decrease in FGF23 was identified in controls after treatment with rapamycin. In rats fed NP, rapamycin decreased mRNA α-klotho/GADPH ratio both in controls, 0.6±0.1 vs 1.1±0.1, p = 0.001, and Nx, 0.3±0.1 vs 0.7±0.1, p = 0.01. At the protein level, a significant reduction in α-klotho was evidenced after treatment with rapamycin both by Western Blot: 0.6±0.1 vs 1.0±0.1, p = 0.01, in controls, 0.7±0.1 vs 1.1±0.1, p = 0.02, in Nx; and by immunohistochemistry staining. Renal α-klotho was inversely correlated with urinary P excretion (r = -0.525, p = 0.0002). The decrease in α-klotho after treatment with rapamycin was also observed in rats fed LP. In conclusion, rapamycin increases phosphaturia and down-regulates α-klotho expression in rats with normal and decreased renal function. These effects can be observed in animals ingesting normal and low P diet.
Collapse
Affiliation(s)
- Azahara Espartero
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Angela Vidal
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Ignacio Lopez
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Ana I. Raya
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Mariano Rodriguez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Escolastico Aguilera-Tejero
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Carmen Pineda
- Department of Animal Medicine and Surgery, University of Cordoba, Campus Universitario Rabanales, Cordoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| |
Collapse
|
15
|
Chen MH, Bai YM, Wu HJ, Li CT, Lin WC, Tsai SJ, Su TP, Tu PC. Role of klotho on antidepressant and antisuicidal effects of low-dose ketamine infusion among patients with treatment-resistant depression and suicidal ideation. J Affect Disord 2023; 340:471-475. [PMID: 37579883 DOI: 10.1016/j.jad.2023.08.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/30/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
OBJECTIVE Previous studies have found an association between klotho, an anti-aging hormone, and major depressive disorder. However, whether low-dose ketamine infusion alters klotho levels among patients with treatment-resistant depression (TRD) remains unknown. METHODS In total, 48 patients with TRD and strong suicidal ideation were randomly assigned to a single 0.5 mg/kg ketamine or 0.045 mg/kg midazolam regimen and were subjected to a 2-week follow-up. Depressive and suicidal symptoms were assessed before the infusion and during the follow-up. The serum levels of klotho were assessed at baseline and 3 days postinfusion. RESULTS A generalized linear model with adjustment of baseline klotho levels showed that, despite the fact that ketamine did not significantly increase levels of klotho, patients in the ketamine group had higher levels of klotho at Day 3 postinfusion than patients in the midazolam group (p = 0.043). However, we found no association between changes in klotho levels and changes in depressive and suicidal symptoms (all p > 0.05). Higher klotho levels at baseline were associated with poorer antidepressant effect of low-dose ketamine during postinfusion follow-up. DISCUSSION Klotho may play a role in the antidepressant effect of low-dose ketamine. Additional molecular studies are necessary to elucidate the neuromechanisms of TRD, ketamine, and klotho.
Collapse
Affiliation(s)
- Mu-Hong Chen
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Ya-Mei Bai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hui-Ju Wu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Cheng-Ta Li
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Chen Lin
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tung-Ping Su
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Psychiatry, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Pei-Chi Tu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
16
|
Sharma K, Zhang G, Hansen J, Bjornstad P, Lee HJ, Menon R, Hejazi L, Liu JJ, Franzone A, Looker HC, Choi BY, Fernandez R, Venkatachalam MA, Kugathasan L, Sridhar VS, Natarajan L, Zhang J, Sharma VS, Kwan B, Waikar SS, Himmelfarb J, Tuttle KR, Kestenbaum B, Fuhrer T, Feldman HI, de Boer IH, Tucci FC, Sedor J, Heerspink HL, Schaub J, Otto EA, Hodgin JB, Kretzler M, Anderton CR, Alexandrov T, Cherney D, Lim SC, Nelson RG, Gelfond J, Iyengar R. Endogenous adenine mediates kidney injury in diabetic models and predicts diabetic kidney disease in patients. J Clin Invest 2023; 133:e170341. [PMID: 37616058 PMCID: PMC10575723 DOI: 10.1172/jci170341] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/10/2023] [Indexed: 08/25/2023] Open
Abstract
Diabetic kidney disease (DKD) can lead to end-stage kidney disease (ESKD) and mortality; however, few mechanistic biomarkers are available for high-risk patients, especially those without macroalbuminuria. Urine from participants with diabetes from the Chronic Renal Insufficiency Cohort (CRIC) study, the Singapore Study of Macro-angiopathy and Micro-vascular Reactivity in Type 2 Diabetes (SMART2D), and the American Indian Study determined whether urine adenine/creatinine ratio (UAdCR) could be a mechanistic biomarker for ESKD. ESKD and mortality were associated with the highest UAdCR tertile in the CRIC study and SMART2D. ESKD was associated with the highest UAdCR tertile in patients without macroalbuminuria in the CRIC study, SMART2D, and the American Indian study. Empagliflozin lowered UAdCR in nonmacroalbuminuric participants. Spatial metabolomics localized adenine to kidney pathology, and single-cell transcriptomics identified ribonucleoprotein biogenesis as a top pathway in proximal tubules of patients without macroalbuminuria, implicating mTOR. Adenine stimulated matrix in tubular cells via mTOR and stimulated mTOR in mouse kidneys. A specific inhibitor of adenine production was found to reduce kidney hypertrophy and kidney injury in diabetic mice. We propose that endogenous adenine may be a causative factor in DKD.
Collapse
Affiliation(s)
- Kumar Sharma
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Guanshi Zhang
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Jens Hansen
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Petter Bjornstad
- Division of Nephrology, Department of Medicine and Section of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Hak Joo Lee
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Rajasree Menon
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Leila Hejazi
- Center for Precision Medicine and
- SygnaMap Inc., San Antonio, Texas, USA
| | - Jian-Jun Liu
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | | | - Helen C. Looker
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| | - Byeong Yeob Choi
- Center for Precision Medicine and
- Department of Population Health Sciences and
| | | | - Manjeri A. Venkatachalam
- Center for Precision Medicine and
- Department of Pathology, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Luxcia Kugathasan
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada. Department of Physiology and Cardiovascular Sciences Collaborative Specialization, University of Toronto, Toronto, Canada
| | - Vikas S. Sridhar
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada. Department of Physiology and Cardiovascular Sciences Collaborative Specialization, University of Toronto, Toronto, Canada
| | - Loki Natarajan
- Herbert Wertheim School of Public Health and
- Moores Cancer Center, University of California, San Diego, La Jolla, California, USA
| | - Jing Zhang
- Moores Cancer Center, University of California, San Diego, La Jolla, California, USA
| | - Varun S. Sharma
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Brian Kwan
- Department of Health Science, California State University, Long Beach, Long Beach, California, USA
| | - Sushrut S. Waikar
- Section of Nephrology, Department of Medicine, Boston Medical Center and Boston University, Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Jonathan Himmelfarb
- Department of Medicine, Division of Nephrology, Kidney Research Institute, University of Washington, Seattle, Washington, USA
| | - Katherine R. Tuttle
- Department of Medicine, Division of Nephrology, Kidney Research Institute, University of Washington, Seattle, Washington, USA
| | - Bryan Kestenbaum
- Department of Medicine, Division of Nephrology, Kidney Research Institute, University of Washington, Seattle, Washington, USA
| | - Tobias Fuhrer
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Harold I. Feldman
- Center for Clinical Epidemiology and Biostatistics and Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA
- Patient-Centered Outcomes Research Institute, Washington, DC, USA
| | - Ian H. de Boer
- Department of Medicine, Division of Nephrology, Kidney Research Institute, University of Washington, Seattle, Washington, USA
| | | | | | - Hiddo Lambers Heerspink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, Netherlands
- The George Institute for Global Health, Sydney, Australia
| | - Jennifer Schaub
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Edgar A. Otto
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jeffrey B. Hodgin
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthias Kretzler
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher R. Anderton
- Center for Precision Medicine and
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Theodore Alexandrov
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - David Cherney
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada. Department of Physiology and Cardiovascular Sciences Collaborative Specialization, University of Toronto, Toronto, Canada
| | - Su Chi Lim
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
- Diabetes Center, Admiralty Medical Center, Khoo Teck Puat Hospital, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Robert G. Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| | - Jonathan Gelfond
- Center for Precision Medicine and
- Department of Population Health Sciences and
| | - Ravi Iyengar
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | |
Collapse
|
17
|
Liu Y, Chen M. Emerging role of α-Klotho in energy metabolism and cardiometabolic diseases. Diabetes Metab Syndr 2023; 17:102854. [PMID: 37722166 DOI: 10.1016/j.dsx.2023.102854] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/16/2023] [Accepted: 09/08/2023] [Indexed: 09/20/2023]
Abstract
BACKGROUND AND AIM Klotho was first identified as a gene associated with aging and longevity in 1997. α-Klotho is an anti-aging protein and its role in energy metabolism, various cardiovascular diseases (CVDs), and metabolic disorders is increasingly being recognized. In this review, we aimed to outline the potential protective role and therapeutic prospects of α-Klotho in energy metabolism and cardiometabolic diseases (CMDs). METHODS We comprehensively reviewed the relevant literature in PubMed using the keywords 'Klotho', 'metabolism', 'cardiovascular', 'diabetes', 'obesity', 'metabolic syndrome', and 'nonalcoholic fatty liver disease'. RESULTS α-Klotho can be divided into membrane-bound Klotho, secreted Klotho, and the most studied circulating soluble Klotho that can act as a hormone. Klotho gene polymorphisms have been implicated in energy metabolism and CMDs. α-Klotho can inhibit insulin/insulin growth factor-1 signaling and its overexpression can lead to a 'healthy insulin resistance' and may exert beneficial effects on the regulation of glycolipid metabolism and central energy homeostasis. α-Klotho, mainly serum Klotho, has been revealed to be protective against CVDs, diabetes and its complications, obesity, and nonalcoholic fatty liver disease. Human recombinant Klotho protein/Klotho gene delivery, multiple drugs, or natural products, and exercise can increase α-Klotho expression. CONCLUSION Overall, α-Klotho has demonstrated its potential as a promising target for modulating energy metabolism and CMDs, and further research is needed to explore its utilization in clinical practice in the future.
Collapse
Affiliation(s)
- Yuanbin Liu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, Hubei, 430000, PR China
| | - Mingkai Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, Hubei, 430000, PR China.
| |
Collapse
|
18
|
Sharma K, Zhang G, Hansen J, Bjornstad P, Lee HJ, Menon R, Hejazi L, Liu JJ, Franzone A, Looker HC, Choi BY, Fernandez R, Venkatachalam MA, Kugathasan L, Sridhar VS, Natarajan L, Zhang J, Sharma V, Kwan B, Waikar S, Himmelfarb J, Tuttle K, Kestenbaum B, Fuhrer T, Feldman H, de Boer IH, Tucci FC, Sedor J, Heerspink HL, Schaub J, Otto E, Hodgin JB, Kretzler M, Anderton C, Alexandrov T, Cherney D, Lim SC, Nelson RG, Gelfond J, Iyengar R. Role of endogenous adenine in kidney failure and mortality with diabetes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.31.23290681. [PMID: 37398187 PMCID: PMC10312877 DOI: 10.1101/2023.05.31.23290681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Diabetic kidney disease (DKD) can lead to end-stage kidney disease (ESKD) and mortality, however, few mechanistic biomarkers are available for high risk patients, especially those without macroalbuminuria. Urine from participants with diabetes from Chronic Renal Insufficiency Cohort (CRIC), Singapore Study of Macro-Angiopathy and Reactivity in Type 2 Diabetes (SMART2D), and the Pima Indian Study determined if urine adenine/creatinine ratio (UAdCR) could be a mechanistic biomarker for ESKD. ESKD and mortality were associated with the highest UAdCR tertile in CRIC (HR 1.57, 1.18, 2.10) and SMART2D (HR 1.77, 1.00, 3.12). ESKD was associated with the highest UAdCR tertile in patients without macroalbuminuria in CRIC (HR 2.36, 1.26, 4.39), SMART2D (HR 2.39, 1.08, 5.29), and Pima Indian study (HR 4.57, CI 1.37-13.34). Empagliflozin lowered UAdCR in non-macroalbuminuric participants. Spatial metabolomics localized adenine to kidney pathology and transcriptomics identified ribonucleoprotein biogenesis as a top pathway in proximal tubules of patients without macroalbuminuria, implicating mammalian target of rapamycin (mTOR). Adenine stimulated matrix in tubular cells via mTOR and stimulated mTOR in mouse kidneys. A specific inhibitor of adenine production was found to reduce kidney hypertrophy and kidney injury in diabetic mice. We propose that endogenous adenine may be a causative factor in DKD.
Collapse
|
19
|
Szőke K, Bódi B, Hendrik Z, Czompa A, Gyöngyösi A, Haines DD, Papp Z, Tósaki Á, Lekli I. Rapamycin treatment increases survival, autophagy biomarkers and expression of the anti-aging klotho protein in elderly mice. Pharmacol Res Perspect 2023; 11:e01091. [PMID: 37190667 DOI: 10.1002/prp2.1091] [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: 03/21/2023] [Accepted: 04/02/2023] [Indexed: 05/17/2023] Open
Abstract
Previous investigations have demonstrated that treatment of animals with rapamycin increases levels of autophagy, which is a process by which cells degrade intracellular detritus, thus suppressing the emergence of senescent cells, whose pro-inflammatory properties, are primary drivers of age-associated physical decline. A hypothesis is tested here that rapamycin treatment of mice approaching the end of their normal lifespan exhibits increased survival, enhanced expression of autophagic proteins; and klotho protein-a biomarker of aging that affects whole organism senescence, and systemic suppression of inflammatory mediator production. Test groups of 24-month-old C57BL mice were injected intraperitoneally with either 1.5 mg/kg/week rapamycin or vehicle. All mice administered rapamycin survived the 12-week course, whereas 43% of the controls died. Relative to controls, rapamycin-treated mice experienced minor but significant weight loss; moreover, nonsignificant trends toward decreased levels of leptin, IL-6, IL-1β, TNF-α, IL-1α, and IGF-1, along with slight elevations in VEGF, MCP-1 were observed in the blood serum of rapamycin-treated mice. Rapamycin-treated mice exhibited significantly enhanced autophagy and elevated expression of klotho protein, particularly in the kidney. Rapamycin treatment also increased cardiomyocyte Ca2+ -sensitivity and enhanced the rate constant of force re-development, which may also contribute to the enhanced survival rate in elderly mice.
Collapse
Affiliation(s)
- Kitti Szőke
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Beáta Bódi
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Hendrik
- Institute of Forensic Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Czompa
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Alexandra Gyöngyösi
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
- Institute of Healthcare Industry, University of Debrecen, Debrecen, Hungary
| | | | - Zoltán Papp
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Árpád Tósaki
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
- ELKH-DE Pharmamodul Research Team, University of Debrecen, Debrecen, Hungary
| | - István Lekli
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
- Institute of Healthcare Industry, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
20
|
Donate-Correa J, Martín-Núñez E, Martin-Olivera A, Mora-Fernández C, Tagua VG, Ferri CM, López-Castillo Á, Delgado-Molinos A, López-Tarruella VC, Arévalo-Gómez MA, Pérez-Delgado N, González-Luis A, Navarro-González JF. Klotho inversely relates with carotid intima- media thickness in atherosclerotic patients with normal renal function (eGFR ≥60 mL/min/1.73m 2): a proof-of-concept study. Front Endocrinol (Lausanne) 2023; 14:1146012. [PMID: 37274332 PMCID: PMC10235765 DOI: 10.3389/fendo.2023.1146012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/26/2023] [Indexed: 06/06/2023] Open
Abstract
INTRODUCTION Klotho protein is predominantly expressed in the kidneys and has also been detected in vascular tissue and peripheral blood circulating cells to a lesser extent. Carotid artery intima-media thickness (CIMT) burden, a marker of subclinical atherosclerosis, has been associated with reductions in circulating Klotho levels in chronic kidney disease patients, who show reduced levels of this protein at all stages of the disease. However, the contribution of serum Klotho and its expression levels in peripheral blood circulating cells and in the carotid artery wall on the CIMT in the absence of kidney impairment has not yet been evaluated. METHODS We conducted a single-center study in 35 atherosclerotic patients with preserved kidney function (eGFR≥60 mL/min/1.73m2) subjected to elective carotid surgery. Serum levels of Klotho and cytokines TNFa, IL6 and IL10 were determined by ELISA and transcripts encoding for Klotho (KL), TNF, IL6 and IL10 from vascular segments were measured by qRT-PCR. Klotho protein expression in the intima-media and adventitia areas was analyzed using immunohistochemistry. RESULTS APatients with higher values of CIMT showed reduced Klotho levels in serum (430.8 [357.7-592.9] vs. 667.8 [632.5-712.9] pg/mL; p<0.001), mRNA expression in blood circulating cells and carotid artery wall (2.92 [2.06-4.8] vs. 3.69 [2.42-7.13] log.a.u., p=0.015; 0.41 [0.16-0.59] vs. 0.79 [0.37-1.4] log.a.u., p=0.013, respectively) and immunoreactivity in the intimal-medial area of the carotids (4.23 [4.15-4.27] vs. 4.49 [4.28-4.63] log µm2 p=0.008). CIMT was inversely related with Klotho levels in serum (r= -0.717, p<0.001), blood mRNA expression (r=-0.426, p=0.011), and with carotid artery mRNA and immunoreactivity levels (r= -0.45, p=0.07; r= -0.455, p= 0.006, respectively). Multivariate analysis showed that serum Klotho, together with the gene expression levels of tumor necrosis factor TNFa in blood circulating cells, were independent determinants of CIMT values (adjusted R2 = 0.593, p<0.001). DISCUSSION The results of this study in subjects with eGFR≥60mL/min/1.73m2 show that patients with carotid artery atherosclerosis and higher values of CIMT present reduced soluble Klotho levels, as well as decreased KL mRNA expression in peripheral blood circulating cells and Klotho protein levels in the intima-media of the carotid artery wall.
Collapse
Affiliation(s)
- Javier Donate-Correa
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria (HUNSC), Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, Santander, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
- RICORS2040 (Red de Investigación Renal-RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
| | - Ernesto Martín-Núñez
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria (HUNSC), Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, Santander, Spain
| | - Alberto Martin-Olivera
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria (HUNSC), Santa Cruz de Tenerife, Spain
| | - Carmen Mora-Fernández
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria (HUNSC), Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, Santander, Spain
- RICORS2040 (Red de Investigación Renal-RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
| | - Víctor G. Tagua
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria (HUNSC), Santa Cruz de Tenerife, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
- Área de Medicina Preventiva y Salud Pública, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Carla M. Ferri
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria (HUNSC), Santa Cruz de Tenerife, Spain
- Escuela de Doctorado y Estudios de Posgrado, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | | | | | | | | | | | - Ainhoa González-Luis
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria (HUNSC), Santa Cruz de Tenerife, Spain
- Escuela de Doctorado y Estudios de Posgrado, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Juan F. Navarro-González
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria (HUNSC), Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, Santander, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
- RICORS2040 (Red de Investigación Renal-RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Nefrología, HUNSC, Santa Cruz de Tenerife, Spain
| |
Collapse
|
21
|
Li SS, Sheng MJ, Sun ZY, Liang Y, Yu LX, Liu QF. Upstream and downstream regulators of Klotho expression in chronic kidney disease. Metabolism 2023; 142:155530. [PMID: 36868370 DOI: 10.1016/j.metabol.2023.155530] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023]
Abstract
Klotho is a critical protein that protects the kidney. Klotho is severely downregulated in chronic kidney disease (CKD), and its deficiency is implicated in the pathogenesis and progression of CKD. Conversely, an increase in Klotho levels results in improved kidney function and delays CKD progression, supporting the notion that modulating Klotho levels could represent a possible therapeutic strategy for CKD treatment. Nevertheless, the regulatory mechanisms responsible for the loss of Klotho remain elusive. Previous studies have demonstrated that oxidative stress, inflammation, and epigenetic modifications can modulate Klotho levels. These mechanisms result in a decrease in Klotho mRNA transcript levels and reduced translation, thus can be grouped together as upstream regulatory mechanisms. However, therapeutic strategies that aim to rescue Klotho levels by targeting these upstream mechanisms do not always result in increased Klotho, indicating the involvement of other regulatory mechanisms. Emerging evidence has shown that endoplasmic reticulum (ER) stress, the unfolded protein response, and ER-associated degradation also affect the modification, translocation, and degradation of Klotho, and thus are proposed to be downstream regulatory mechanisms. Here, we discuss the current understanding of upstream and downstream regulatory mechanisms of Klotho and examine potential therapeutic strategies to upregulate Klotho expression for CKD treatment.
Collapse
Affiliation(s)
- Sha-Sha Li
- Clinical Research & Lab Centre, Affiliated Kunshan Hospital of Jiangsu University, 91 Qianjin West Road, Kunshan, Jiangsu 215300, China
| | - Ming-Jie Sheng
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, 91 Qianjin West Road, Kunshan, Jiangsu 215300, China
| | - Zhuo-Yi Sun
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, 91 Qianjin West Road, Kunshan, Jiangsu 215300, China
| | - Yan Liang
- Gusu School, Nanjing Medical University, The First People's Hospital of Kunshan, 91 Qianjin West Road, Kunshan, Jiangsu 215300, China
| | - Li-Xia Yu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, 91 Qianjin West Road, Kunshan, Jiangsu 215300, China.
| | - Qi-Feng Liu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, 91 Qianjin West Road, Kunshan, Jiangsu 215300, China; Gusu School, Nanjing Medical University, The First People's Hospital of Kunshan, 91 Qianjin West Road, Kunshan, Jiangsu 215300, China.
| |
Collapse
|
22
|
Donate-Correa J, Matos-Perdomo E, González-Luis A, Martín-Olivera A, Ortiz A, Mora-Fernández C, Navarro-González JF. The Value of Klotho in Kidney Transplantation. Transplantation 2023; 107:616-627. [PMID: 36253904 DOI: 10.1097/tp.0000000000004331] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Kidney transplant recipients have better survival rates and improved quality of life than long-term dialysis patients. However, delayed graft function, immunosuppressive therapy nephrotoxicity, and rejection episodes may compromise graft and patient survival. The KL gene is highly expressed in kidney tubular cells and encodes the antiaging and kidney-protective protein Klotho, which has membrane-anchored and soluble forms and regulates mineral metabolism. Klotho expression decreases during acute kidney injury or chronic kidney disease, and human chronic kidney disease shares features of accelerated aging with murine Klotho deficiency. In this work, we review clinical studies on the relationship between Klotho and kidney transplantation. Specifically, we address the dynamics of serum and kidney Klotho levels in donors and kidney transplant recipients, the role of Klotho as a marker of current graft function and graft outcomes, and the potential impact of Klotho on kidney protection in the transplantation context. A better understanding of the potential biomarker and therapeutic utility of Klotho in kidney transplant recipients may provide new insights into the control of graft function and new therapeutic strategies to preserve allograft function.
Collapse
Affiliation(s)
- Javier Donate-Correa
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, Santander, Spain
- Instituto de Tecnologías Biomédicas, University of La Laguna, Santa Cruz de Tenerife, Spain
| | - Emiliano Matos-Perdomo
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Doctoral and Graduate School, University of La Laguna, San Cristóbal de La Laguna, Tenerife, Spain
| | - Ainhoa González-Luis
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Instituto de Tecnologías Biomédicas, University of La Laguna, Santa Cruz de Tenerife, Spain
- Doctoral and Graduate School, University of La Laguna, San Cristóbal de La Laguna, Tenerife, Spain
| | - Alberto Martín-Olivera
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Instituto de Tecnologías Biomédicas, University of La Laguna, Santa Cruz de Tenerife, Spain
- Doctoral and Graduate School, University of La Laguna, San Cristóbal de La Laguna, Tenerife, Spain
| | - Alberto Ortiz
- Instituto de Investigación Sanitaria Fundación Jiménez-Díaz-Universidad Autónoma de Madrid, Madrid, Spain
- RICORS2040 (Red de Investigación Renal-RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Mora-Fernández
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, Santander, Spain
- RICORS2040 (Red de Investigación Renal-RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan F Navarro-González
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, Santander, Spain
- Instituto de Tecnologías Biomédicas, University of La Laguna, Santa Cruz de Tenerife, Spain
- RICORS2040 (Red de Investigación Renal-RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
23
|
The Anti-Aging Hormone Klotho Promotes Retinal Pigment Epithelium Cell Viability and Metabolism by Activating the AMPK/PGC-1α Pathway. Antioxidants (Basel) 2023; 12:antiox12020385. [PMID: 36829944 PMCID: PMC9952846 DOI: 10.3390/antiox12020385] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/08/2023] Open
Abstract
Initially discovered by Makuto Kuro-o in 1997, Klotho is a putative aging-suppressor gene when overexpressed and accelerates aging when deleted in mice. Previously, we showed that α-Klotho regulates retinal pigment epithelium (RPE) functions and protects against oxidative stress. However, the mechanisms by which Klotho influences RPE and retinal homeostasis remain elusive. Here, by performing a series of in vitro and in vivo experiments, we demonstrate that Klotho regulates cell viability under oxidative stress, mitochondrial gene expression and activity by inducing the phosphorylation of AMPK and p38MAPK, which in turn phosphorylate and activate CREB and ATF2, respectively, triggering PGC-1α transcription. The inhibition of Klotho in human RPE cells using CRISPR-Cas9 gene editing confirmed that a lack of Klotho negatively affects RPE functions, including mitochondrial activity and cell viability. Proteomic analyses showed that myelin sheath and mitochondrial-related proteins are downregulated in the RPE/retina of Kl-/- compared to WT mice, further supporting our biochemical observations. We conclude that Klotho acts upstream of the AMPK/PGC-1α pathway and regulates RPE/retinal resistance to oxidative stress, mitochondrial function, and gene and protein expressions. Thus, KL decline during aging could negatively impact retinal health, inducing age-related retinal degeneration.
Collapse
|
24
|
Pan W, Jie W, Huang H. Vascular calcification: Molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2023; 4:e200. [PMID: 36620697 PMCID: PMC9811665 DOI: 10.1002/mco2.200] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 01/05/2023] Open
Abstract
Vascular calcification (VC) is recognized as a pathological vascular disorder associated with various diseases, such as atherosclerosis, hypertension, aortic valve stenosis, coronary artery disease, diabetes mellitus, as well as chronic kidney disease. Therefore, it is a life-threatening state for human health. There were several studies targeting mechanisms of VC that revealed the importance of vascular smooth muscle cells transdifferentiating, phosphorous and calcium milieu, as well as matrix vesicles on the progress of VC. However, the underlying molecular mechanisms of VC need to be elucidated. Though there is no acknowledged effective therapeutic strategy to reverse or cure VC clinically, recent evidence has proved that VC is not a passive irreversible comorbidity but an active process regulated by many factors. Some available approaches targeting the underlying molecular mechanism provide promising prospects for the therapy of VC. This review aims to summarize the novel findings on molecular mechanisms and therapeutic interventions of VC, including the role of inflammatory responses, endoplasmic reticulum stress, mitochondrial dysfunction, iron homeostasis, metabolic imbalance, and some related signaling pathways on VC progression. We also conclude some recent studies on controversial interventions in the clinical practice of VC, such as calcium channel blockers, renin-angiotensin system inhibitions, statins, bisphosphonates, denosumab, vitamins, and ion conditioning agents.
Collapse
Affiliation(s)
- Wei Pan
- Department of Cardiology, the Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Joint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic DiseaseSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Wei Jie
- Department of Cardiology, the Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Joint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic DiseaseSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Hui Huang
- Department of Cardiology, the Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Joint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic DiseaseSun Yat‐sen UniversityShenzhenGuangdongChina
| |
Collapse
|
25
|
Tang A, Zhang Y, Wu L, Lin Y, Lv L, Zhao L, Xu B, Huang Y, Li M. Klotho's impact on diabetic nephropathy and its emerging connection to diabetic retinopathy. Front Endocrinol (Lausanne) 2023; 14:1180169. [PMID: 37143722 PMCID: PMC10151763 DOI: 10.3389/fendo.2023.1180169] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 04/04/2023] [Indexed: 05/06/2023] Open
Abstract
Diabetic nephropathy (DN) is the leading cause of end-stage renal disease worldwide and is a significant burden on healthcare systems. α-klotho (klotho) is a protein known for its anti-aging properties and has been shown to delay the onset of age-related diseases. Soluble klotho is produced by cleavage of the full-length transmembrane protein by a disintegrin and metalloproteases, and it exerts various physiological effects by circulating throughout the body. In type 2 diabetes and its complications DN, a significant decrease in klotho expression has been observed. This reduction in klotho levels may indicate the progression of DN and suggest that klotho may be involved in multiple pathological mechanisms that contribute to the onset and development of DN. This article examines the potential of soluble klotho as a therapeutic agent for DN, with a focus on its ability to impact multiple pathways. These pathways include anti-inflammatory and oxidative stress, anti-fibrotic, endothelial protection, prevention of vascular calcification, regulation of metabolism, maintenance of calcium and phosphate homeostasis, and regulation of cell fate through modulation of autophagy, apoptosis, and pyroptosis pathways. Diabetic retinopathy shares similar pathological mechanisms with DN, and targeting klotho may offer new insights into the prevention and treatment of both conditions. Finally, this review assesses the potential of various drugs used in clinical practice to modulate klotho levels through different mechanisms and their potential to improve DN by impacting klotho levels.
Collapse
Affiliation(s)
- Anqi Tang
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Yu Zhang
- Department of Nephrology, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi’an, China
| | - Ling Wu
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Yong Lin
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Lizeyu Lv
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Liangbin Zhao
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Bojun Xu
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Youqun Huang
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Mingquan Li
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
- *Correspondence: Mingquan Li,
| |
Collapse
|
26
|
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: 22] [Impact Index Per Article: 7.3] [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
|
27
|
Dutta P, Sengupta A, Chakraborty S. Epigenetics: a new warrior against cardiovascular calcification, a forerunner in modern lifestyle diseases. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:62093-62110. [PMID: 34601672 DOI: 10.1007/s11356-021-15718-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/26/2021] [Indexed: 06/13/2023]
Abstract
Arterial and aortic valve calcifications are the most prevalent pathophysiological conditions among all the reported cases of cardiovascular calcifications. It increases with several risk factors like age, hypertension, external stimuli, mechanical forces, lipid deposition, malfunction of genes and signaling pathways, enhancement of naturally occurring calcium inhibitors, and many others. Modern-day lifestyle is affected by numerous environmental factors and harmful toxins that impair our health rather than providing benefits. Applying the combinatorial approach or targeting the exact mechanism could be a new strategy for drug designing or attenuating the severity of calcification. Most of the non-communicable diseases are life-threatening; thus, altering the phenotype and not the genotype may reveal the gateway for fighting with upcoming hurdles. Overall, this review summarizes the reason behind the generation of arterial and aortic valve calcification and its related signaling pathways and also the detrimental effects of calcification. In addition, the individual process of epigenetics and how the implementation of this process becomes a novel approach for diminishing the harmful effect of calcification are discussed. Noteworthy, as epigenetics is linked with genetics and environmental factors necessitates further clinical trials for complete and in-depth understanding and application of this strategy in a more specific and prudent manner.
Collapse
Affiliation(s)
- Parna Dutta
- Department of Life Sciences, Presidency University, 86/1, College Street, Baker building, 2nd floor, Kolkata, West Bengal, 700073, India
| | - Arunima Sengupta
- Department of Life science & Bio-technology, Jadavpur University, Kolkata, 700032, India
| | - Santanu Chakraborty
- Department of Life Sciences, Presidency University, 86/1, College Street, Baker building, 2nd floor, Kolkata, West Bengal, 700073, India.
| |
Collapse
|
28
|
Prud’homme GJ, Kurt M, Wang Q. Pathobiology of the Klotho Antiaging Protein and Therapeutic Considerations. FRONTIERS IN AGING 2022; 3:931331. [PMID: 35903083 PMCID: PMC9314780 DOI: 10.3389/fragi.2022.931331] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/06/2022] [Indexed: 12/06/2022]
Abstract
The α-Klotho protein (henceforth denoted Klotho) has antiaging properties, as first observed in mice homozygous for a hypomorphic Klotho gene (kl/kl). These mice have a shortened lifespan, stunted growth, renal disease, hyperphosphatemia, hypercalcemia, vascular calcification, cardiac hypertrophy, hypertension, pulmonary disease, cognitive impairment, multi-organ atrophy and fibrosis. Overexpression of Klotho has opposite effects, extending lifespan. In humans, Klotho levels decline with age, chronic kidney disease, diabetes, Alzheimer’s disease and other conditions. Low Klotho levels correlate with an increase in the death rate from all causes. Klotho acts either as an obligate coreceptor for fibroblast growth factor 23 (FGF23), or as a soluble pleiotropic endocrine hormone (s-Klotho). It is mainly produced in the kidneys, but also in the brain, pancreas and other tissues. On renal tubular-cell membranes, it associates with FGF receptors to bind FGF23. Produced in bones, FGF23 regulates renal excretion of phosphate (phosphaturic effect) and vitamin D metabolism. Lack of Klotho or FGF23 results in hyperphosphatemia and hypervitaminosis D. With age, human renal function often deteriorates, lowering Klotho levels. This appears to promote age-related pathology. Remarkably, Klotho inhibits four pathways that have been linked to aging in various ways: Transforming growth factor β (TGF-β), insulin-like growth factor 1 (IGF-1), Wnt and NF-κB. These can induce cellular senescence, apoptosis, inflammation, immune dysfunction, fibrosis and neoplasia. Furthermore, Klotho increases cell-protective antioxidant enzymes through Nrf2 and FoxO. In accord, preclinical Klotho therapy ameliorated renal, cardiovascular, diabetes-related and neurodegenerative diseases, as well as cancer. s-Klotho protein injection was effective, but requires further investigation. Several drugs enhance circulating Klotho levels, and some cross the blood-brain barrier to potentially act in the brain. In clinical trials, increased Klotho was noted with renin-angiotensin system inhibitors (losartan, valsartan), a statin (fluvastatin), mTOR inhibitors (rapamycin, everolimus), vitamin D and pentoxifylline. In preclinical work, antidiabetic drugs (metformin, GLP-1-based, GABA, PPAR-γ agonists) also enhanced Klotho. Several traditional medicines and/or nutraceuticals increased Klotho in rodents, including astaxanthin, curcumin, ginseng, ligustilide and resveratrol. Notably, exercise and sport activity increased Klotho. This review addresses molecular, physiological and therapeutic aspects of Klotho.
Collapse
Affiliation(s)
- Gérald J. Prud’homme
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada
- *Correspondence: Gérald J. Prud’homme,
| | - Mervé Kurt
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China
- Shanghai Yinuo Pharmaceutical Co., Ltd., Shanghai, China
| |
Collapse
|
29
|
Kim HJ, Kim Y, Kang M, Kim S, Park SK, Sung S, Hyun YY, Jung JY, Ahn C, Oh KH. Low Klotho/Fibroblast Growth Factor 23 Ratio Is an Independent Risk Factor for Renal Progression in Chronic Kidney Disease: Finding From KNOW-CKD. Front Med (Lausanne) 2022; 9:904963. [PMID: 35872753 PMCID: PMC9304693 DOI: 10.3389/fmed.2022.904963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 06/07/2022] [Indexed: 11/21/2022] Open
Abstract
Background We aimed to evaluate soluble Klotho and circulating fibroblast growth factor 23 (FGF23) ratio as a risk factor for renal progression, cardiovascular (CV) events, and mortality in chronic kidney disease (CKD). Methods We analyzed 2,099 subjects from a CKD cohort whose soluble Klotho and C-terminal FGF23 levels were measured at enrollment. The Klotho to FGF23 ratio was calculated as Klotho values divided by FGF23 values + 1 (hereinafter called the Klotho/FGF23 ratio). Participants were categorized into quartiles according to Klotho/FGF23 ratio. The primary outcome was renal events, defined as the doubling of serum creatinine, 50% reduction of estimated glomerular filtration rate from the baseline values, or development of end-stage kidney disease. The secondary outcomes consisted of CV events and death. Changes in CV parameters at the time of enrollment and during follow-up according to the Klotho/FGF23 ratio were also examined. Results During the follow-up period of 64.0 ± 28.2 months, 735 (35.1%) and 273 (13.0%) subjects developed renal events and composite outcomes of CV events and death, respectively. After adjustment, the first (HR: 1.36; 95% CI: 1.08–1.72, P = 0.010) and second (HR: 1.45; 95% CI: 1.15–1.83, P = 0.002) quartiles with regard to the Klotho/FGF23 ratio showed elevated risk of renal events as compared to the fourth quartile group. There was no significant association between Klotho/FGF23 ratio and the composite outcome of CV events and death. The prevalence of left ventricular hypertrophy and vascular calcification was higher in the low Klotho/FGF23 ratio quartiles at baseline and at the fourth-year follow-up. Conclusions Low Klotho/FGF23 ratio was significantly associated with increased renal events in the cohort of Korean predialysis CKD patients.
Collapse
Affiliation(s)
- Hyo Jin Kim
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, South Korea
- Biomedical Research Institute, Pusan National University Hospital, Busan, South Korea
| | - Yunmi Kim
- Department of Internal Medicine, Inje University Busan Paik Hospital, Busan, South Korea
| | - Minjung Kang
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Seonmi Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Sue Kyung Park
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Suah Sung
- Department of Internal Medicine, Eulji Medical Center, Eulji University, Seoul, South Korea
| | - Young Youl Hyun
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Ji Yong Jung
- Department of Internal Medicine, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, South Korea
| | - Curie Ahn
- Department of Internal Medicine, National Medical Center, Seoul, South Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
- *Correspondence: Kook-Hwan Oh
| |
Collapse
|
30
|
Xu JP, Zeng RX, He MH, Lin SS, Guo LH, Zhang MZ. Associations Between Serum Soluble α-Klotho and the Prevalence of Specific Cardiovascular Disease. Front Cardiovasc Med 2022; 9:899307. [PMID: 35795366 PMCID: PMC9251131 DOI: 10.3389/fcvm.2022.899307] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/20/2022] [Indexed: 12/03/2022] Open
Abstract
Objective Accumulating experimental evidence has identified the beneficial effects of the anti-aging protein, serum soluble α-Klotho, on longevity, and the cardiovascular system. Although a previous study has revealed the predictive value of α-Klotho on total cardiovascular disease (CVD), the associations between α-Klotho and specific CVDs, including congestive heart failure (CHF), coronary heart disease (CHD), myocardial infarction (MI), and stroke, remains to be fully elucidated in humans. Methods For 8,615 adults in the 2007 to 2016 National Health and Nutrition Examination Survey, stratified multivariable logistic regression models, restricted cubic spline curves, and subgroup analyses were used to evaluate the associations between α-Klotho and the four specific CVDs. Results In the quartile analyses, compared to those in the highest quartile, participants in the lowest level of α-Klotho were significantly associated with CHF [odds ratio (OR) = 1.46, 95% CI: 1.09–1.97] and MI (1.33, 1.02–1.74), which was not the case for CHD (1.12, 0.91–1.38) or stroke (0.96, 0.73–1.25). Each unit increment in the ln-transformed α-Klotho concentrations was only positively associated with a 38 and 24% reduction in the prevalence of CHF and MI, respectively. Restricted cubic spline curves indicated that the α-Klotho was correlated with CHF and MI in linear-inverse relationships. Conclusion The present findings suggested that the serum soluble α-Klotho is significantly associated with the prevalence of CHF and MI. To better determine whether α-Klotho is a specific biomarker of CVD, particularly for CHD and stroke, further research in humans is needed.
Collapse
Affiliation(s)
- Jun-Peng Xu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Rui-Xiang Zeng
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Mu-Hua He
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shan-Shan Lin
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Li-Heng Guo
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Min-Zhou Zhang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- *Correspondence: Min-Zhou Zhang
| |
Collapse
|
31
|
Rao Z, Zheng Y, Xu L, Wang Z, Zhou Y, Chen M, Dong N, Cai Z, Li F. Endoplasmic Reticulum Stress and Pathogenesis of Vascular Calcification. Front Cardiovasc Med 2022; 9:918056. [PMID: 35783850 PMCID: PMC9243238 DOI: 10.3389/fcvm.2022.918056] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/30/2022] [Indexed: 12/05/2022] Open
Abstract
Vascular calcification (VC) is characterized by calcium phosphate deposition in blood vessel walls and is associated with many diseases, as well as increased cardiovascular morbidity and mortality. However, the molecular mechanisms underlying of VC development and pathogenesis are not fully understood, thus impeding the design of molecular-targeted therapy for VC. Recently, several studies have shown that endoplasmic reticulum (ER) stress can exacerbate VC. The ER is an intracellular membranous organelle involved in the synthesis, folding, maturation, and post-translational modification of secretory and transmembrane proteins. ER stress (ERS) occurs when unfolded/misfolded proteins accumulate after a disturbance in the ER environment. Therefore, downregulation of pathological ERS may attenuate VC. This review summarizes the relationship between ERS and VC, focusing on how ERS regulates the development of VC by promoting osteogenic transformation, inflammation, autophagy, and apoptosis, with particular interest in the molecular mechanisms occurring in various vascular cells. We also discuss, the therapeutic effects of ERS inhibition on the progress of diseases associated with VC are detailed.
Collapse
Affiliation(s)
- Zhenqi Rao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yidan Zheng
- Basic Medical School, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihao Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhejun Cai
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
32
|
Martín-Núñez E, Pérez-Castro A, Tagua VG, Hernández-Carballo C, Ferri C, Pérez-Delgado N, Rodríguez-Ramos S, Cerro-López P, López-Castillo Á, Delgado-Molinos A, López-Tarruella VC, Arévalo-Gómez MA, González-Luis A, Martín-Olivera A, Morales-Estévez CC, Mora-Fernández C, Donate-Correa J, Navarro-González JF. Klotho expression in peripheral blood circulating cells is associated with vascular and systemic inflammation in atherosclerotic vascular disease. Sci Rep 2022; 12:8422. [PMID: 35590090 PMCID: PMC9120199 DOI: 10.1038/s41598-022-12548-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/04/2022] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide. New therapeutic strategies are aimed to modulate the athero-inflammatory process that partially orchestrates underlying vascular damage. Peripheral blood circulating cells include different immune cells with a central role in the development of the atherogenic inflammatory response. The anti-aging protein α-Klotho has been related to protective effects against CVD. KL is expressed in monocytes, macrophages, and lymphocytes where it exerts anti-inflammatory effects. In this work, we analyse the relationships of the levels of inflammatory markers with the expression of the KL gene in PBCCs and with the serum levels of soluble KL in atherosclerotic vascular disease. For this, we conducted a cross-sectional single-center case-control study including a study group of 76 CVD patients and a control group of 16 cadaveric organ donors without medical antecedent or study indicating CVD. Vascular artery fragments and whole blood and serum samples were obtained during elective or organ retrieval surgery. Serum levels of sKL, TNFα and IL10, and gene expression levels of KL, TNF, IL10, NFKB1, DNMT1, and DNMT3A in PBCCs were measured. In these cells, we also determined KL promoter methylation percentage. Histological and immunohistochemical analyses were employed to visualize atherosclerotic lesions and to measure IL10 and TNFα levels in vascular fragments. Patients with CVD presented higher values of proinflammatory markers both at systemic and in the vasculature and in the PBCCs, compared to the control group. In PBCCs, CVD patients also presented lower gene expression levels of KL gene (56.4% difference, P < 0.001), higher gene expression levels of DNMT1 and DNMT3A (P < 0.0001, for both) and a higher methylation status of in the promoter region of KL (34.1 ± 4.1% vs. 14.6 ± 3.4%, P < 0.01). In PBCCs and vasculature, KL gene expression correlated inversely with pro-inflammatory markers and directly with anti-inflammatory markers. sKL serum levels presented similar associations with the expression levels of pro- and anti-inflammatory markers in PBCCs. The differences in KL expression levels in PBCCs and in serum sKL levels with respect to control group was even greater in those CVD patients with macroscopically observable atheromatous plaques. We conclude that promoter methylation-mediated downregulation of KL gene expression in PBCCs is associated with the pro-inflammatory status in atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Ernesto Martín-Núñez
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain
- Escuela de Doctorado Y Estudios de Posgrado, Universidad de La Laguna, 38200, San Cristóbal de La Laguna, Tenerife, Spain
| | - Atteneri Pérez-Castro
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain
- Escuela de Doctorado Y Estudios de Posgrado, Universidad de La Laguna, 38200, San Cristóbal de La Laguna, Tenerife, Spain
| | - Víctor G Tagua
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain
- Instituto Universitario de Enfermedades Tropicales Y Salud Pública de Canarias, Universidad de La Laguna, 38200, San Cristóbal de La Laguna, Tenerife, Spain
| | - Carolina Hernández-Carballo
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain
- Escuela de Doctorado Y Estudios de Posgrado, Universidad de La Laguna, 38200, San Cristóbal de La Laguna, Tenerife, Spain
| | - Carla Ferri
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain
- Escuela de Doctorado Y Estudios de Posgrado, Universidad de La Laguna, 38200, San Cristóbal de La Laguna, Tenerife, Spain
| | - Nayra Pérez-Delgado
- Servicio de Análisis Clínicos, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain
| | - Sergio Rodríguez-Ramos
- Coordinación de Trasplantes, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain
| | - Purificación Cerro-López
- Coordinación de Trasplantes, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain
| | - Ángel López-Castillo
- Servicio de Cirugía Vascular, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain
| | - Alejandro Delgado-Molinos
- Servicio de Cirugía Vascular, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain
| | - Victoria Castro López-Tarruella
- Servicio de Anatomía Patológica, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain
| | - Miguel A Arévalo-Gómez
- Departamento de Anatomía E Histología Humana, Universidad de Salamanca, 37008, Salamanca, Spain
| | - Ainhoa González-Luis
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain
- Escuela de Doctorado Y Estudios de Posgrado, Universidad de La Laguna, 38200, San Cristóbal de La Laguna, Tenerife, Spain
| | - Alberto Martín-Olivera
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain
| | | | - Carmen Mora-Fernández
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain
| | - Javier Donate-Correa
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain.
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, 38200, San Cristóbal de La Laguna, Tenerife, Spain.
| | - Juan F Navarro-González
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain.
- Servicio de Nefrología, Hospital Universitario Nuestra Señora de Candelaria, 38010, Santa Cruz de Tenerife, Tenerife, Spain.
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, 38200, San Cristóbal de La Laguna, Tenerife, Spain.
| |
Collapse
|
33
|
Etemadi J, Samadifar M, Ghojazadeh M, Motavalli R, Oriyo R, Majidi T, Tayebi Khosroshahi H. The Effects of Cholecalciferol Supplementation on FGF23 and α-Klotho in Hemodialysis Patients With Hypovitaminosis D: A Randomized, Double-Blind, Placebo-Controlled Trial. J Ren Nutr 2022; 32:334-340. [PMID: 34294550 DOI: 10.1053/j.jrn.2021.05.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/24/2021] [Accepted: 05/25/2021] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVE Vitamin D-fibroblast growth factor-23 (FGF-23)-klotho forms an axis that takes part at least in cardiovascular complications in patients with chronic kidney disease. This study aimed to assess the effects of cholecalciferol supplementation on FGF23 and α-klotho in patients with hypovitaminosis D requiring hemodialysis. METHODS In a single-center, parallel-arm, randomized, double-blind, placebo-controlled trial, 86 patients with hypovitaminosis D requiring hemodialysis were enrolled. The patients were randomized into 2 groups (n = 43 each) to receive either 50,000 IU of cholecalciferol or placebo every week for 12 weeks. Accordingly, the serum levels of FGF23 and klotho were measured by ELISA and compared between both groups. RESULTS Serum 25OH(D) levels increased in participants who received cholecalciferol supplementation compared with participants who received placebo (P = .006). In addition, serum FGF23 decreased and α-klotho levels increased in the supplemented group compared with placebo. However, the before-after differences between cholecalciferol supplement and placebo were significant only for α-klotho (P = .035). These effects were not accompanied by changes in the levels of phosphate, total and ionized calcium, and intact parathyroid hormone. CONCLUSION Cholecalciferol supplementation of 50,000 IU for 12 weeks increases α-klotho levels in the serum of kidney failure patients undergoing hemodialysis. This may suggest that patients receiving maintenance hemodialysis can benefit from using cholecalciferol supplementation and increase in serum α-klotho levels.
Collapse
Affiliation(s)
- Jalal Etemadi
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Samadifar
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Ghojazadeh
- Research Center for Evidence based-medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roza Motavalli
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roghaiyeh Oriyo
- Imam Reza Teaching Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Taraneh Majidi
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
34
|
Arroyo E, Troutman AD, Moorthi RN, Avin KG, Coggan AR, Lim K. Klotho: An Emerging Factor With Ergogenic Potential. FRONTIERS IN REHABILITATION SCIENCES 2022; 2:807123. [PMID: 36188832 PMCID: PMC9397700 DOI: 10.3389/fresc.2021.807123] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/10/2021] [Indexed: 11/13/2022]
Abstract
Sarcopenia and impaired cardiorespiratory fitness are commonly observed in older individuals and patients with chronic kidney disease (CKD). Declines in skeletal muscle function and aerobic capacity can progress into impaired physical function and inability to perform activities of daily living. Physical function is highly associated with important clinical outcomes such as hospitalization, functional independence, quality of life, and mortality. While lifestyle modifications such as exercise and dietary interventions have been shown to prevent and reverse declines in physical function, the utility of these treatment strategies is limited by poor widespread adoption and adherence due to a wide variety of both perceived and actual barriers to exercise. Therefore, identifying novel treatment targets to manage physical function decline is critically important. Klotho, a remarkable protein with powerful anti-aging properties has recently been investigated for its role in musculoskeletal health and physical function. Klotho is involved in several key processes that regulate skeletal muscle function, such as muscle regeneration, mitochondrial biogenesis, endothelial function, oxidative stress, and inflammation. This is particularly important for older adults and patients with CKD, which are known states of Klotho deficiency. Emerging data support the existence of Klotho-related benefits to exercise and for potential Klotho-based therapeutic interventions for the treatment of sarcopenia and its progression to physical disability. However, significant gaps in our understanding of Klotho must first be overcome before we can consider its potential ergogenic benefits. These advances will be critical to establish the optimal approach to future Klotho-based interventional trials and to determine if Klotho can regulate physical dysfunction.
Collapse
Affiliation(s)
- Eliott Arroyo
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ashley D. Troutman
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University Purdue University, Indianapolis, IN, United States
| | - Ranjani N. Moorthi
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Keith G. Avin
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University Purdue University, Indianapolis, IN, United States
| | - Andrew R. Coggan
- Department of Kinesiology, School of Health and Human Sciences, Indiana University Purdue University Indianapolis, Indianapolis, IN, United States
| | - Kenneth Lim
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
35
|
Franco ML, Beyerstedt S, Rangel ÉB. Klotho and Mesenchymal Stem Cells: A Review on Cell and Gene Therapy for Chronic Kidney Disease and Acute Kidney Disease. Pharmaceutics 2021; 14:11. [PMID: 35056905 PMCID: PMC8778857 DOI: 10.3390/pharmaceutics14010011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease (CKD) and acute kidney injury (AKI) are public health problems, and their prevalence rates have increased with the aging of the population. They are associated with the presence of comorbidities, in particular diabetes mellitus and hypertension, resulting in a high financial burden for the health system. Studies have indicated Klotho as a promising therapeutic approach for these conditions. Klotho reduces inflammation, oxidative stress and fibrosis and counter-regulates the renin-angiotensin-aldosterone system. In CKD and AKI, Klotho expression is downregulated from early stages and correlates with disease progression. Therefore, the restoration of its levels, through exogenous or endogenous pathways, has renoprotective effects. An important strategy for administering Klotho is through mesenchymal stem cells (MSCs). In summary, this review comprises in vitro and in vivo studies on the therapeutic potential of Klotho for the treatment of CKD and AKI through the administration of MSCs.
Collapse
Affiliation(s)
- Marcella Liciani Franco
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil; (M.L.F.); (S.B.)
| | - Stephany Beyerstedt
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil; (M.L.F.); (S.B.)
| | - Érika Bevilaqua Rangel
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil; (M.L.F.); (S.B.)
- Nephrology Division, Federal University of São Paulo, Sao Paulo 04038-901, Brazil
| |
Collapse
|
36
|
Liu Q, Yu L, Yin X, Ye J, Li S. Correlation Between Soluble Klotho and Vascular Calcification in Chronic Kidney Disease: A Meta-Analysis and Systematic Review. Front Physiol 2021; 12:711904. [PMID: 34483963 PMCID: PMC8414804 DOI: 10.3389/fphys.2021.711904] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/14/2021] [Indexed: 11/29/2022] Open
Abstract
Background: The correlation between soluble Klotho (sKlotho) level and vascular calcification (VC) in patients with chronic kidney disease (CKD) remains controversial. Using meta-analysis, we aimed to address this controversy and assess the feasibility of applying sKlotho as a biomarker for VC. Methods: Medical electronic databases were thoroughly searched for eligible publications on the association between sKlotho level and VC in CKD patients. Effectors, including correlation coefficients (r), odds ratios (ORs), hazard ratio (HR) or β-values, and 95% confidence intervals (CIs) were extracted and combined according to study design or effector calculation method. Pooled effectors were generated using both random-effects models and fixed-effects models according to I 2-value. Origin of heterogeneity was explored by sensitivity analysis and subgroup analysis. Results: Ten studies with 1,204 participants from a total of 1,199 publications were eligible and included in this meta-analysis. The combined correlation coefficient (r) was [-0.33 (-0.62, -0.04)] with significant heterogeneity (I 2 = 89%, p < 0.001) based on Spearman correlation analysis, and this significant association was also demonstrated in subgroups. There was no evidence of publication bias. The combined OR was [3.27 (1.70, 6.30)] with no evidence of heterogeneity (I 2 = 0%, p = 0.48) when sKlotho was treated as a categorical variable or [1.05 (1.01, 1.09)] with moderate heterogeneity (I 2 = 63%, p = 0.10) when sKlotho was treated as a continuous variable based on multivariate logistic regression. No significant association was observed and the pooled OR was [0.29 (0.01, 11.15)] with high heterogeneity (I 2 = 96%, p < 0.001) according to multivariate linear regression analysis. There was an inverse association between sKlotho and parathyroid hormone levels. The combined coefficient (r) was [-0.20 (-0.40, -0.01)] with significant heterogeneity (I 2 = 86%, p < 0.001), and without obvious publication bias. No significant association was found between sKlotho and calcium or phosphate levels. Conclusion: There exists a significant association between decreased sKlotho level and increased risk of VC in CKD patients. This raises the possibility of applying sKlotho as a biomarker for VC in CKD populations. Large, prospective, well-designed studies or interventional clinical trials are required to validate our findings.
Collapse
Affiliation(s)
- QiFeng Liu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - LiXia Yu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - XiaoYa Yin
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - JianMing Ye
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - ShaSha Li
- Clinical Research & Lab Centre, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| |
Collapse
|
37
|
Tang PK, Geddes RF, Jepson RE, Elliott J. A feline-focused review of chronic kidney disease-mineral and bone disorders - Part 2: Pathophysiology of calcium disorder and extraosseous calcification. Vet J 2021; 275:105718. [PMID: 34329743 DOI: 10.1016/j.tvjl.2021.105718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 06/23/2021] [Accepted: 07/21/2021] [Indexed: 10/20/2022]
Abstract
Derangements in mineral metabolism are one of the main entities in chronic kidney disease-mineral and bone disorder (CKD-MBD). This is the second of a two-part review of the physiology and pathophysiology of calcium homeostasis in feline CKD-MBD. While dysregulation in calcium homeostasis is known to contribute to the development of vascular calcification in CKD, evidence characterising the relationship between serum calcium concentration and nephrocalcinosis and nephrolithiasis is limited. Recently, fibroblast growth factor 23 (FGF23) and α-Klotho have gained increased research interest and been shown to be important biomarkers for the prediction of CKD progression in human patients. However, conflicting evidence exists on their role in calcium homeostasis and vascular and soft tissue calcification. This review details the pathophysiology of calcium disorders associated with CKD-MBD and its implications on vascular and soft tissue mineralisation in human and feline patients. Further prospective studies investigating the clinical consequences of calcium disturbances in cats with CKD are warranted and this may provide additional insight into the pathophysiology of feline CKD-MBD.
Collapse
Affiliation(s)
- Pak-Kan Tang
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London, United Kingdom.
| | - Rebecca F Geddes
- Department of Clinical Science and Services, Royal Veterinary College, University of London, London, United Kingdom
| | - Rosanne E Jepson
- Department of Clinical Science and Services, Royal Veterinary College, University of London, London, United Kingdom
| | - Jonathan Elliott
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London, United Kingdom
| |
Collapse
|
38
|
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
|
39
|
Bhat OM, Yuan X, Kukreja RC, Li PL. Regulatory role of mammalian target of rapamycin signaling in exosome secretion and osteogenic changes in smooth muscle cells lacking acid ceramidase gene. FASEB J 2021; 35:e21732. [PMID: 34143450 PMCID: PMC8221173 DOI: 10.1096/fj.202100385r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/22/2021] [Accepted: 06/01/2021] [Indexed: 12/28/2022]
Abstract
Acid ceramidase (murine gene code: Asah1) (50 kDa) belongs to N-terminal nucleophile hydrolase family. This enzyme is located in the lysosome, which mediates conversion of ceramide (CER) into sphingosine and free fatty acids at acidic pH. CER plays an important role in intracellular sphingolipid metabolism and its increase causes inflammation. The mammalian target of rapamycin complex 1 (mTORC1) signaling on late endosomes (LEs)/lysosomes may control cargo selection, membrane biogenesis, and exosome secretion, which may be fine controlled by lysosomal sphingolipids such as CER. This lysosomal-CER-mTOR signaling may be a crucial molecular mechanism responsible for development of arterial medial calcification (AMC). Torin-1 (5 mg/kg/day), an mTOR inhibitor, significantly decreased aortic medial calcification accompanied with decreased expression of osteogenic markers like osteopontin (OSP) and runt-related transcription factor 2 (RUNX2) and upregulation of smooth muscle 22α (SM22-α) in mice receiving high dose of Vitamin D (500 000 IU/kg/day). Asah1fl/fl /SMCre mice had markedly increased co-localization of mTORC1 with lysosome-associated membrane protein-1 (Lamp-1) (lysosome marker) and decreased co-localization of vacuolar protein sorting-associated protein 16 (VPS16) (a multivesicular bodies [MVBs] marker) with Lamp-1, suggesting mTOR activation caused reduced MVBs interaction with lysosomes. Torin-1 significantly reduced the co-localization of mTOR vs Lamp-1, increased lysosome-MVB interaction which was associated with reduced accumulation of CD63 and annexin 2 (exosome markers) in the coronary arterial wall of mice. Using coronary artery smooth muscle cells (CASMCs), Pi -stimulation significantly increased p-mTOR expression in Asah1fl/fl /SMCre CASMCs as compared to WT/WT cells associated with increased calcium deposition and mineralization. Torin-1 blocked Pi -induced calcium deposition and mineralization. siRNA mTOR and Torin-1 significantly reduce co-localization of mTORC1 with Lamp-1, increased VPS16 vs Lamp-1 co-localization in Pi -stimulated CASMCs, associated with decreased exosome release. Functionally, Torin-1 significantly reduces arterial stiffening as shown by restoration from increased pulse wave velocity and decreased elastin breaks. These results suggest that lysosomal CER-mTOR signaling may play a critical role for the control of lysosome-MVB interaction, exosome secretion and arterial stiffening during AMC.
Collapse
Affiliation(s)
- Owais M. Bhat
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298
| | - Xinxu Yuan
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298
| | - Rakesh C. Kukreja
- VCU Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, 1101 East Marshall Street, Richmond, VA 23298-0204
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298
| |
Collapse
|
40
|
Cai H, Zhu X, Lu J, Zhu M, Liu S, Zhan Y, Ni Z, Gu L, Zhang W, Mou S. A Decreased Level of Soluble Klotho Can Predict Cardiovascular Death in No or Mild Abdominal Aortic Calcification Hemodialysis Patients. Front Med (Lausanne) 2021; 8:672000. [PMID: 34079811 PMCID: PMC8165200 DOI: 10.3389/fmed.2021.672000] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/22/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Soluble Klotho plays an important role in cardiovascular disease and death in chronic kidney disease (CKD). We assessed the relationship between serum soluble Klotho (sKL) level and outcome in MHD patients. Methods: Soluble Klotho was detected by ELISA. Cox regression analysis and Kaplan-Meier analysis showed the relationship between sKL and cardiovascular disease (CVD) mortality in maintenance hemodialysis (MHD) patients. Results: There were 45 cases (35.2%) of all-cause death and 36 cases (28.1%) of CVD mortality. Multivariate linear regression analysis showed that Log[iPTH] (γ = −0.224, P = 0.015) was an independent predictor of sKL level. Cox regression showed that lower sKL was associated with higher CVD mortality rate [OR = 0.401, 95% CI (0.183–0.867), P = 0.022]. Kaplan-Meier analysis showed that the CVD mortality rate increased significantly in patients with low sKL (P = 0.006). Compared with high sKL patients, low sKL patients with no or mild vascular calcification [aortic calcification score (AACs) ≤ 4] had no significant difference in all-cause mortality rate. The CVD mortality rate was significantly lower in high sKL patients (P = 0.004) than in those with low sKL. In the severe calcification group (AACs ≥ 5), all-cause and CVD mortality rates were similar between different sKL groups (P = 0.706 and 0.488, respectively). The area under the receiver-operating characteristic curve (AUC) of soluble Klotho for predicting the CVD in MHD patients with AACs ≤ 4 was 0.796 (0.647–0.946, P = 0.017), sensitivity was 0.921, and specificity was 0.50 for a cutoff value of 307.69 pg/ml. Conclusions: Lower sKL was associated with higher CVD mortality rate. Lower sKL concentration in MHD patients with no or mild calcification can predict CVD mortality.
Collapse
Affiliation(s)
- Hong Cai
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xuying Zhu
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jiayue Lu
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Minxia Zhu
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shang Liu
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yaping Zhan
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhaohui Ni
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Leyi Gu
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Weiming Zhang
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shan Mou
- Department of Nephrology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
41
|
Lu Y, Yuan T, Min X, Yuan Z, Cai Z. AMPK: Potential Therapeutic Target for Vascular Calcification. Front Cardiovasc Med 2021; 8:670222. [PMID: 34046440 PMCID: PMC8144331 DOI: 10.3389/fcvm.2021.670222] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/08/2021] [Indexed: 12/18/2022] Open
Abstract
Vascular calcification (VC) is an urgent worldwide health issue with no available medical treatment. It is an active cell-driven process by osteogenic differentiation of vascular cells with complex mechanisms. The AMP-activated protein kinase (AMPK) serves as the master sensor of cellular energy status. Accumulating evidence reveals the vital role of AMPK in VC progression. AMPK is involved in VC in various ways, including inhibiting runt-related transcription factor 2 signaling pathways, triggering autophagy, attenuating endoplasmic reticulum stress and dynamic-related protein 1-mediated mitochondrial fission, and activating endothelial nitric oxide synthase. AMPK activators, like metformin, are associated with reduced calcification deposits in certain groups of patients, indicating that AMPK is a potential therapeutic target for VC.
Collapse
Affiliation(s)
- Yi Lu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tan Yuan
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinjia Min
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhen Yuan
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhejun Cai
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Jiaxing Key Laboratory of Cardiac Rehabilitation, Jiaxing, China
| |
Collapse
|
42
|
Clemens Z, Sivakumar S, Pius A, Sahu A, Shinde S, Mamiya H, Luketich N, Cui J, Dixit P, Hoeck JD, Kreuz S, Franti M, Barchowsky A, Ambrosio F. The biphasic and age-dependent impact of klotho on hallmarks of aging and skeletal muscle function. eLife 2021; 10:e61138. [PMID: 33876724 PMCID: PMC8118657 DOI: 10.7554/elife.61138] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 04/06/2021] [Indexed: 12/15/2022] Open
Abstract
Aging is accompanied by disrupted information flow, resulting from accumulation of molecular mistakes. These mistakes ultimately give rise to debilitating disorders including skeletal muscle wasting, or sarcopenia. To derive a global metric of growing 'disorderliness' of aging muscle, we employed a statistical physics approach to estimate the state parameter, entropy, as a function of genes associated with hallmarks of aging. Escalating network entropy reached an inflection point at old age, while structural and functional alterations progressed into oldest-old age. To probe the potential for restoration of molecular 'order' and reversal of the sarcopenic phenotype, we systemically overexpressed the longevity protein, Klotho, via AAV. Klotho overexpression modulated genes representing all hallmarks of aging in old and oldest-old mice, but pathway enrichment revealed directions of changes were, for many genes, age-dependent. Functional improvements were also age-dependent. Klotho improved strength in old mice, but failed to induce benefits beyond the entropic tipping point.
Collapse
Affiliation(s)
- Zachary Clemens
- Department of Physical Medicine & Rehabilitation, University of PittsburghPittsburghUnited States
- Department of Environmental and Occupational Health, University of PittsburghPittsburghUnited States
| | - Sruthi Sivakumar
- Department of Physical Medicine & Rehabilitation, University of PittsburghPittsburghUnited States
- Department of Bioengineering, University of PittsburghPittsburghUnited States
| | - Abish Pius
- Department of Physical Medicine & Rehabilitation, University of PittsburghPittsburghUnited States
- Department of Computational & Systems Biology, School of Medicine, University of PittsburghPittsburghUnited States
| | - Amrita Sahu
- Department of Physical Medicine & Rehabilitation, University of PittsburghPittsburghUnited States
| | - Sunita Shinde
- Department of Physical Medicine & Rehabilitation, University of PittsburghPittsburghUnited States
| | - Hikaru Mamiya
- Department of Bioengineering, University of PittsburghPittsburghUnited States
| | - Nathaniel Luketich
- Department of Bioengineering, University of PittsburghPittsburghUnited States
| | - Jian Cui
- Department of Computational & Systems Biology, School of Medicine, University of PittsburghPittsburghUnited States
| | - Purushottam Dixit
- Department of Physics, University of FloridaGainesvilleUnited States
| | - Joerg D Hoeck
- Department of Research Beyond Borders, Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals, IncRheinGermany
| | - Sebastian Kreuz
- Department of Research Beyond Borders, Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals, IncRheinGermany
| | - Michael Franti
- Department of Research Beyond Borders, Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals, IncRheinGermany
| | - Aaron Barchowsky
- Department of Environmental and Occupational Health, University of PittsburghPittsburghUnited States
| | - Fabrisia Ambrosio
- Department of Physical Medicine & Rehabilitation, University of PittsburghPittsburghUnited States
- Department of Environmental and Occupational Health, University of PittsburghPittsburghUnited States
- Department of Bioengineering, University of PittsburghPittsburghUnited States
- McGowan Institute for Regenerative Medicine, University of PittsburghPittsburghUnited States
| |
Collapse
|
43
|
Podestà MA, Cucchiari D, Ciceri P, Messa P, Torregrosa JV, Cozzolino M. Cardiovascular calcifications in kidney transplant recipients. Nephrol Dial Transplant 2021; 37:2063-2071. [PMID: 33620476 DOI: 10.1093/ndt/gfab053] [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: 01/05/2021] [Indexed: 12/15/2022] Open
Abstract
Vascular and valvular calcifications are highly prevalent in kidney transplant recipients and are associated with an increased risk of cardiovascular events, which represent the leading cause of long-term mortality in these patients. However, cardiovascular calcification has been traditionally considered as a condition mostly associated with advanced chronic kidney disease stages and dialysis, and comparatively fewer studies have assessed its impact after kidney transplantation. Despite partial or complete resolution of uremia-associated metabolic derangements, kidney transplant recipients are still exposed to several pro-calcifying stimuli that favour the progression of pre-existing vascular calcifications or their de novo development. Traditional risk factors, bone mineral disorders, inflammation, immunosuppressive drugs and deficiency of calcification inhibitors may all play a role, and strategies to correct or minimize their effects are urgently needed. The aim of this work is to provide an overview of established and putative mediators involved in the pathogenesis of cardiovascular calcification in kidney transplantation, and to describe the clinical and radiological features of these forms. We also discuss current evidence on preventive strategies to delay the progression of cardiovascular calcifications in kidney transplant recipients, as well as novel therapeutic candidates to potentially prevent their long-term deleterious effects.
Collapse
Affiliation(s)
- Manuel Alfredo Podestà
- Renal Division, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Italy
| | - David Cucchiari
- Nephrology and Renal Transplant Department, Hospital Clínic, Barcelona, Spain
| | - Paola Ciceri
- Department of Nephrology, Dialysis and Renal Transplant, Renal Research Laboratory, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Piergiorgio Messa
- Department of Nephrology, Dialysis and Renal Transplant, Renal Research Laboratory, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Mario Cozzolino
- Renal Division, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Italy
| |
Collapse
|
44
|
Serum soluble Klotho is inversely related to coronary artery calcification assessed by intravascular ultrasound in patients with stable coronary artery disease. J Cardiol 2020; 77:583-589. [PMID: 33303310 DOI: 10.1016/j.jjcc.2020.11.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/09/2020] [Accepted: 11/19/2020] [Indexed: 01/31/2023]
Abstract
BACKGROUND Although the Klotho gene is recognized as an aging-suppressor gene, the clinical significance of its soluble product, soluble Klotho, in coronary artery disease (CAD) has not been completely determined. The relationship between soluble Klotho and coronary artery calcification (CAC) was investigated in patients with stable CAD. METHODS CAC in culprit lesions was analyzed in 75 non-dialysis patients with stable CAD who were scheduled for percutaneous coronary intervention (PCI) following intravascular ultrasound (IVUS). The main outcome measure was the calcium index (CalcIndex), a volumetric IVUS-derived measure of total calcification per culprit lesion. A low CalcIndex was defined as a first-quartile calcium index (<0.042). Patients were divided into two groups according to the median serum Klotho value: low Klotho (n = 37, ≤460 pg/mL) and high Klotho (n = 38, >460 pg/mL). RESULTS The CalcIndex was significantly lower in patients with high than with low Klotho. Patients with high Klotho had a significantly higher prevalence of a low CalcIndex than those with low Klotho. The number of angiographic moderate-severe CACs in whole coronary arteries was significantly decreased in patients with high Klotho compared to low Klotho. Serum Klotho levels correlated significantly and inversely with the CalcIndex. This relationship was pronounced in patients with estimated glomerular filtration rate <60 mL/min/1.73 m2. Logistic regression analysis showed that high Klotho was associated with a low CalcIndex independent of classical coronary risk factors and markers of mineral metabolism. CONCLUSIONS High serum soluble Klotho levels are associated with a low degree of CAC in non-dialysis, stable CAD patients treated by PCI.
Collapse
|
45
|
Neutel CHG, Hendrickx JO, Martinet W, De Meyer GRY, Guns PJ. The Protective Effects of the Autophagic and Lysosomal Machinery in Vascular and Valvular Calcification: A Systematic Review. Int J Mol Sci 2020; 21:E8933. [PMID: 33255685 PMCID: PMC7728070 DOI: 10.3390/ijms21238933] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/19/2020] [Accepted: 11/21/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Autophagy is a highly conserved catabolic homeostatic process, crucial for cell survival. It has been shown that autophagy can modulate different cardiovascular pathologies, including vascular calcification (VCN). OBJECTIVE To assess how modulation of autophagy, either through induction or inhibition, affects vascular and valvular calcification and to determine the therapeutic applicability of inducing autophagy. DATA SOURCES A systematic review of English language articles using MEDLINE/PubMed, Web of Science (WoS) and the Cochrane library. The search terms included autophagy, autolysosome, mitophagy, endoplasmic reticulum (ER)-phagy, lysosomal, calcification and calcinosis. Study characteristics: Thirty-seven articles were selected based on pre-defined eligibility criteria. Thirty-three studies (89%) studied vascular smooth muscle cell (VSMC) calcification of which 27 (82%) studies investigated autophagy and six (18%) studies lysosomal function in VCN. Four studies (11%) studied aortic valve calcification (AVCN). Thirty-four studies were published in the time period 2015-2020 (92%). CONCLUSION There is compelling evidence that both autophagy and lysosomal function are critical regulators of VCN, which opens new perspectives for treatment strategies. However, there are still challenges to overcome, such as the development of more selective pharmacological agents and standardization of methods to measure autophagic flux.
Collapse
Affiliation(s)
| | | | | | | | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, University of Antwerp, 2610 Antwerp, Belgium; (C.H.G.N.); (J.O.H.); (W.M.); (G.R.Y.D.M.)
| |
Collapse
|
46
|
Inverse sex-based expression profiles of PTEN and Klotho in mice. Sci Rep 2020; 10:20189. [PMID: 33214645 PMCID: PMC7677532 DOI: 10.1038/s41598-020-77217-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 10/30/2020] [Indexed: 01/08/2023] Open
Abstract
Sex differences are considered predictive factors in the development of several neurological diseases, which are also known to coincide with impaired phosphoinositide 3-kinase (PI3K)-AKT pathway activity, an essential signaling cascade involved in the control of several cellular functions such as autophagy and apoptosis. Here, under physiological conditions, we show important sex differences in the underlying balancing mechanisms that lead to similar AKT activity levels and autophagy and apoptosis processes in the two sexes. We demonstrate inverse sex-based expression of PTEN and Klotho, two important proteins that are known to negatively regulate the AKT pathway, and inverse sex-dependent levels of mTOR and FoxO3a activity. Taken together, our findings indicate that inverse sex-based regulation may be one of the underlying balancing mechanisms that differ between the sexes and a possible cause of sex-based autophagic and apoptotic responses to triggering situations that can lead to a sex-based predisposition to some neurological diseases.
Collapse
|
47
|
Daneshgar N, Dai DF. ROS, Klotho and mTOR in cardiorenal aging. Aging (Albany NY) 2020; 12:19830-19831. [PMID: 33125344 PMCID: PMC7655185 DOI: 10.18632/aging.104209] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 06/11/2023]
Affiliation(s)
- Nastaran Daneshgar
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Dao-Fu Dai
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| |
Collapse
|
48
|
Huang Y, Wang J, Luo M, Yan D, Zhang C. Carnosine attenuates vascular smooth muscle cells calcification through mTOR signaling pathway. Aging Med (Milton) 2020; 3:153-158. [PMID: 33103035 PMCID: PMC7574631 DOI: 10.1002/agm2.12125] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 08/23/2020] [Accepted: 08/23/2020] [Indexed: 01/10/2023] Open
Abstract
Objective Vascular calcification is prevalent in the aging population, as we know that arterial calcification is associated with aging. Recent studies have demonstrated that carnosine, a naturally occurring dipeptide, performs the treatment of aging‐related diseases, such as atherosclerosis and type 2 diabetes. Here, we investigated the role of carnosine in a calcification model of vascular smooth muscle cells (VSMCs). Methods In this research, we used an in vitro model of VSMC calcification to investigate the role of carnosine in the progression of rat VSMC calcification. Results Carnosine treatment attenuated calcium deposition in a dose‐dependent manner, detected by Alizarin Red S staining and calcium content assay. Carnosine also reduced the protein level of Runx2, bone morphogenetic protein 2 (BMP‐2), and cellular reactive oxygen species (ROS) production. Further, carnosine inhibited the activation of the mammalian target of rapamycin (mTOR) pathway. Conclusion Carnosine attenuated the VSMC calcification via inhibition of osteoblastic transdifferentiation and the mTOR signaling pathway.
Collapse
Affiliation(s)
- Yi Huang
- Department of Geriatrics Institute of Gerontology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jinli Wang
- Department of Geriatrics Institute of Gerontology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Mandi Luo
- Department of Geriatrics Institute of Gerontology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Dan Yan
- Department of Geriatrics Institute of Gerontology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Cuntai Zhang
- Department of Geriatrics Institute of Gerontology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
49
|
Zheng J, Zhu X, He Y, Hou S, Liu T, Zhi K, Hou T, Gao L. CircCDK8 regulates osteogenic differentiation and apoptosis of PDLSCs by inducing ER stress/autophagy during hypoxia. Ann N Y Acad Sci 2020; 1485:56-70. [PMID: 32978798 DOI: 10.1111/nyas.14483] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/28/2020] [Accepted: 08/11/2020] [Indexed: 12/17/2022]
Abstract
Mounting evidence indicates that circular RNAs (circRNAs) have essential roles in several diseases, including periodontitis. Periodontal ligament stem cells (PDLSCs) exhibit potential for treating periodontitis accompanied by hypoxia. However, it is unclear how circRNA affects the osteogenesis of PDLSCs under hypoxia. In this study, a novel circRNA, hsa_circ_0003489, was found located at the gene for cyclin-dependent kinase 8 (CDK8) and referred to as circCDK8. The expression levels of circCDK8 and hypoxia-inducible factor-1α were significantly increased in periodontitis tissues, and the expression of circCDK8 was further confirmed in a hypoxia model using cobalt chloride (CoCl2 ). Interestingly, the results showed that the expression levels of osteoblast markers (RUNX2, ALP, OCN, and COL1A1) were increased in CoCl2 -treated PDLSCs at 6 and 12 h, but decreased at 24, 48, and 72 h. On the basis of bioinformatics and functional experiments, CoCl2 also induced endoplasmic reticulum stress, autophagy, and apoptosis of PDLSCs; the inhibition of autophagy promoted the osteogenic differentiation of CoCl2 -treated PDLSCs. Furthermore, circCDK8 overexpression induced autophagy and apoptosis through mTOR signaling, and circCDK8 silencing reversed the inhibitory effects of CoCl2 on osteogenic differentiation of PDLSCs. In conclusion, our results indicate that circCDK8 represses the osteogenic differentiation of PDLSCs by triggering autophagy activation in a hypoxic microenvironment. CircCDK8 could be a new therapeutic target of periodontitis.
Collapse
Affiliation(s)
- Jingjing Zheng
- Department of Endodontics, Key Laboratory of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ximei Zhu
- Department of Endodontics, Stomatological Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Yani He
- Department of Endodontics, Stomatological Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Siyu Hou
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Ting Liu
- Department of Endodontics, Key Laboratory of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Keqian Zhi
- Department of Oral and Maxillofacial Surgery, Key Laboratory of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Tiezhou Hou
- Department of Endodontics, Stomatological Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Ling Gao
- Department of Oral and Maxillofacial Surgery, Key Laboratory of Oral Clinical Medicine, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
50
|
Buchanan S, Combet E, Stenvinkel P, Shiels PG. Klotho, Aging, and the Failing Kidney. Front Endocrinol (Lausanne) 2020; 11:560. [PMID: 32982966 PMCID: PMC7481361 DOI: 10.3389/fendo.2020.00560] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/09/2020] [Indexed: 12/11/2022] Open
Abstract
Klotho has been recognized as a gene involved in the aging process in mammals for over 30 years, where it regulates phosphate homeostasis and the activity of members of the fibroblast growth factor (FGF) family. The α-Klotho protein is the receptor for Fibroblast Growth Factor-23 (FGF23), regulating phosphate homeostasis and vitamin D metabolism. Phosphate toxicity is a hallmark of mammalian aging and correlates with diminution of Klotho levels with increasing age. As such, modulation of Klotho activity is an attractive target for therapeutic intervention in the diseasome of aging; in particular for chronic kidney disease (CKD), where Klotho has been implicated directly in the pathophysiology. A range of senotherapeutic strategies have been developed to directly or indirectly influence Klotho expression, with varying degrees of success. These include administration of exogenous Klotho, synthetic and natural Klotho agonists and indirect approaches, via modulation of the foodome and the gut microbiota. All these approaches have significant potential to mitigate loss of physiological function and resilience accompanying old age and to improve outcomes within the diseasome of aging.
Collapse
Affiliation(s)
- Sarah Buchanan
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Emilie Combet
- School of Medicine, Dentistry & Nursing, Human Nutrition, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Peter Stenvinkel
- Division of Renal Medicine M99, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Paul G. Shiels
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|