1
|
Wang K, Liu J. Anti-aging protein α-Klotho is potential for reducing comorbidity risk of cardiometabolic diseases in vulnerable populations and enhancing long-term prognosis. Sci Rep 2025; 15:16722. [PMID: 40369033 PMCID: PMC12078659 DOI: 10.1038/s41598-025-01580-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 05/07/2025] [Indexed: 05/16/2025] Open
Abstract
This study investigated the impact of anti-aging protein α-Klotho on cardiometabolic diseases (CMDs) among middle-aged and elderly population. A total of 11,198 participants aged 40-79 years were included in the National Health and Nutrition Examination Survey (NHANES) spanning 2007-2016. Serum α-Klotho levels were quantified via enzyme-linked immunosorbent assays. CMDs comprised cardiovascular disease (CVD), and four metabolic disorders: type 2 diabetes (T2DM), obesity, chronic kidney disease (CKD), and non-alcoholic fatty liver disease (NAFLD). Weighted logistic regression analysis, subgroup analysis, mediation analysis, restricted cubic splines (RCS), and Cox proportional hazards regression analysis were used. α-Klotho exhibited negative associations with each single CMD except T2DM, and RCS showed U-shape and L-shape dose-response relationships of α-Klotho with risk of T2DM and CKD, respectively. Ordered logistic regression analysis revealed that higher levels of Klotho markedly reduced the cumulative number of metabolic comorbidities complicating CVD (OR 0.56 (0.35, 0.91)). Simple mediation analysis showed CKD may explain up to 20.42% of the association between Klotho and CVD. Notably, α-Klotho's association with cardiometabolic comorbidities was particularly evident among individuals who were widowed/divorced/separated, non-Hispanic Black, lower-income, or less educated, with hypertension, current smokers, lower leisure and commuting physical activity, but higher work-related physical activity. Regarding long-term effects, higher α-Klotho levels were associated with lower all-cause mortality among participants with CMDs, but not among those without CMDs. Higher α-Klotho levels were associated with lower CMD prevalence, particularly in high-risk cardiovascular populations with lower socioeconomic status and unfavorable lifestyles and reduced all-cause mortality risk among CMD patients.
Collapse
Affiliation(s)
- Kai Wang
- Medical School, Southeast University, Nanjing, China
| | - Jianing Liu
- Medical Faculty, Ulm University, Ulm, Germany.
| |
Collapse
|
2
|
Capasso G, Franssen CFM, Perna AF, Massy ZA, Menzies RI, Zoccali C, Tessitore A, Nedergaard M, Okusa MD, Ortiz A, Wagner CA, Unwin RJ. Drivers and mechanisms of cognitive decline in chronic kidney disease. Nat Rev Nephrol 2025:10.1038/s41581-025-00963-0. [PMID: 40281076 DOI: 10.1038/s41581-025-00963-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 04/29/2025]
Abstract
Cognitive impairment is highly prevalent among individuals with chronic kidney disease (CKD). Despite its high prevalence, the contributing factors and mechanisms underlying brain-kidney dysfunction in CKD remain poorly understood. However, advances in neuroscience, including novel imaging techniques and cognitive assessment methods, have begun to clarify this complex relationship. Several factors contribute directly to cognitive decline in people with CKD, including accumulation of uraemic toxins, microvascular damage, malnutrition, chronic inflammation and disruptions in key neuroprotective pathways, such as those involving Klotho and the glymphatic system. These factors are also linked to the accelerated ageing observed in people with CKD, a key contributor to cognitive decline. However, most studies on cognition in people with CKD have been cross-sectional and associative, offering limited insight into causation. Research advances, such as studies on the effect of uraemic toxins on the blood-brain barrier and the role of the endothelial glycocalyx in vascular damage, offer promising new directions. Emerging data from longitudinal cohort studies are also enhancing our understanding of these processes, with potential implications for both the treatment of CKD-related cognitive decline and the broader issue of cognitive dysfunction in ageing populations. Here, we examine key mechanisms linking CKD to cognitive decline and consider potential therapeutic interventions.
Collapse
Affiliation(s)
- Giovambattista Capasso
- Biogem, Institute of Molecular Biology and Genetics, Ariano Irpino, Italy.
- Department of Translational Medical Science, University of Campania Luigi Vanvitelli, Naples, Italy.
| | - Casper F M Franssen
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Alessandra F Perna
- Department of Translational Medical Science, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ziad A Massy
- AURA (Association pour l'Utilization du Rein Artificiel dans la Region Parisienne) Paris, Department of Nephrology, CHU Ambroise Paré, AP-HP, Paris, France
- Centre for Research in Epidemiology and Population Health (CESP), University Paris-Saclay, University Versailles-Saint Quentin, Inserm UMRS 1018, Clinical Epidemiology Team, Villejuif, France
| | - Robert I Menzies
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Carmine Zoccali
- Biogem, Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale (IPNET), c/o Nefrologia, Grande Ospedale Metropolitano, Reggio, Italy
| | - Alessandro Tessitore
- Department of Advanced Surgical and Medical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Maiken Nedergaard
- Center for Basic and Translational Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Mark D Okusa
- Division of Nephrology, Center for Immunity Inflammation and Regenerative Medicine University of Virginia, Charlottesville, VA, USA
| | - Alberto Ortiz
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD UAM), Madrid, Spain
| | - Carsten A Wagner
- Institute of Physiology and Zurich Kidney Center University of Zurich, Zurich, Switzerland
| | - Robert J Unwin
- UCL Centre for Kidney and Bladder Health, University College London, London, UK
| |
Collapse
|
3
|
Liu X, Zhao J, Liu J, Huang Y, Deng W, Yan L, Cui M, Pan X, Xiao H, Liu X. Association of α-Klotho with anti-aging effects of Ganoderma lucidum in animal models. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119597. [PMID: 40057146 DOI: 10.1016/j.jep.2025.119597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/15/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aging is a complex, universal process characterized by structural and functional decline across multiple organs. Ganoderma lucidum (G. lucidum), a renowned traditional Chinese medicinal fungus, has long been recognized for its anti-aging properties. However, the underlying mechanisms remain incompletely understood. AIM OF THE STUDY This study aimed to investigate the anti-aging effects of G. lucidum and its underlying mechanisms. MATERIALS AND METHODS We investigated the anti-aging effects of G. lucidum sporoderm-broken spore powder (Gl-SBSP) on Caenorhabditis elegans (C. elegans) lifespan and aging across multiple organs using natural aging, D-galactose (D-gal)-induced aging, and radiation-induced premature senescence mouse models. In C. elegans, we assessed lifespan, reproductive capacity, body length, pharyngeal pumping, body bends, fat and lipofuscin levels, as well as reactive oxygen species (ROS) accumulation. In mice, histopathological staining, complete blood counts, and enzyme-linked immunosorbent assay (ELISA) were used to evaluate tissue damage, while quantitative real-time PCR (RT-qPCR) was employed to access small intestine barrier integrity. Western blot (WB) and immunohistochemistry (IHC) were utilized to analyze the distribution of alpha Klotho (α-Klotho) in the kidney, blood, and urine. RESULTS Gl-SBSP significantly extended C. elegans lifespan, improved reproductive capacity and mobility, and reduced lipofuscin and ROS levels. In naturally aged mice, Gl-SBSP enhanced physical appearance and performance. Additionally, Gl-SBSP alleviated aging-related structural and functional decline in multiple organs, including the colon, spleen, kidneys, liver, and small intestine, across all aging models. Biochemical analyses revealed that Gl-SBSP increased transmembrane α-Klotho (mα-Klotho) and soluble α-Klotho (sα-Klotho) levels in kidney tissue and elevated sα-Klotho levels in serum and urine. CONCLUSION This study is the first to demonstrate that G. lucidum exerts α-Klotho-associated anti-aging effects in animal models, highlighting its potential as an anti-aging intervention.
Collapse
Affiliation(s)
- Xiaojing Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Jiamin Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Jia Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Yan Huang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Wei Deng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Luwen Yan
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin, 300192, China
| | - Xinhua Pan
- Jiangxi Xiankelai Biotechnology Co., Ltd., 10 Shacheng Road, Jiujiang, 332000, China
| | - Huiwen Xiao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Xingzhong Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| |
Collapse
|
4
|
Li H, Ouyang Y, Lv H, Liang H, Luo S, Zhang Y, Mao H, Chen T, Chen W, Zhou Y, Liu Q. Nanoparticle-mediated Klotho gene therapy prevents acute kidney injury to chronic kidney disease transition through regulating PPARα signaling in renal tubular epithelial cells. Biomaterials 2025; 315:122926. [PMID: 39500111 DOI: 10.1016/j.biomaterials.2024.122926] [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: 04/08/2024] [Revised: 09/05/2024] [Accepted: 10/27/2024] [Indexed: 12/09/2024]
Abstract
Klotho is an anti-aging protein produced primarily by tubular epithelial cells (TECs). Down-regulated expression of Klotho in injured TECs plays a key pathogenic role in promoting acute kidney injury (AKI) to chronic kidney disease (CKD) transition, yet therapeutic approaches targeting the restoration of renal Klotho levels remain challenging for clinical application. Here, we synthesize polydopamine-polyethylenimine-l-serine-Klotho plasmid nanoparticles (PPSK NPs), which can safely and selectively deliver the Klotho gene to the injured TECs through binding kidney injury molecule-1 and maintain the expression of Klotho protein. In vitro, PPSK NPs effectively reduce the hypoxia-reoxygenation-induced reactive oxygen species production and fibrotic gene expression. In the unilateral ischemia-reperfusion injury- and folic acid-induced AKI-CKD transition mouse models, a single low-dose injection of PPSK NPs is sufficient to preserve the normal kidney architecture and prevent renal fibrosis. Mechanismly, the protective effect of PPSK NPs relies on upregulating a key molecule peroxisome proliferator-activated receptor alpha (PPARα) via the inhibition of p38 and JNK phosphorylation, which in turn improves tubular fatty acid beta-oxidation and reduces renal lipid accumulation, thereby protecting against kidney fibrosis. In conclusion, our results highlight the translational potential of nanoparticle-based Klotho gene therapy in preventing the AKI-CKD transition.
Collapse
Affiliation(s)
- Hongyu Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Yuying Ouyang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Haoran Lv
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Hanzhi Liang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Siweier Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China; Basic and Translational Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yating Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China; Basic and Translational Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Haiping Mao
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Tianfeng Chen
- Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China.
| | - Yiming Zhou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China; Basic and Translational Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| | - Qinghua Liu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China; Department of Nephrology, Jieyang People's Hospital, Jieyang, 522000, China.
| |
Collapse
|
5
|
Grigore TV, Leusink QM, Zuidscherwoude M, Bos C, Olauson H, Hoenderop J. Partial renal deletion of Klotho is not sufficient to impact renal electrolyte handling in distal convoluted tubule specific knock-out mice. Physiol Rep 2025; 13:e70297. [PMID: 40165603 PMCID: PMC11959153 DOI: 10.14814/phy2.70297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
Klotho controls renal electrolyte handling by modulating tubular reabsorption of calcium and phosphate through the epithelial calcium channel TRPV5 and sodium phosphate co-transporter NPT2A. The Ksp-KL-/- mice have a targeted deletion of Klotho in the distal part of the nephron. Considering that the distal convoluted tubule is the most important site for Ca2+ regulation in the kidney, Ksp-KL-/- mice were challenged with a Ca2+-deficient diet for determining the Ca2+ handling and pinpointing the Klotho levels needed for controlling renal Ca2+ handling. The Ksp-KL-/- mice displayed normal weight and showed unaltered calcium and phosphate levels in serum and 24-h urine. Expression of calciotropic (Trpv5, Trpv6) and phosphotropic (Slc34a1, Slc34a2) genes in the kidneys, duodenum, ileum, and colon were not affected by Klotho deletion. In conclusion, our study reports that mice with 18%-93% residual levels of Klotho in the kidney exhibit normal electrolyte homeostasis when placed on a low Ca2+-content diet.
Collapse
Affiliation(s)
- Teodora V. Grigore
- Department of Medical BiosciencesResearch Institute for Medical Innovation, Radboud University Medical CenterNijmegenThe Netherlands
| | - Quinty M. Leusink
- Department of Medical BiosciencesResearch Institute for Medical Innovation, Radboud University Medical CenterNijmegenThe Netherlands
| | - Malou Zuidscherwoude
- Department of Medical BiosciencesResearch Institute for Medical Innovation, Radboud University Medical CenterNijmegenThe Netherlands
| | - Caro Bos
- Department of Medical BiosciencesResearch Institute for Medical Innovation, Radboud University Medical CenterNijmegenThe Netherlands
| | - Hannes Olauson
- Division of Renal MedicineDepartment of Clinical Science, Intervention and Technology, Karolinska InstitutetStockholmSweden
| | - Joost Hoenderop
- Department of Medical BiosciencesResearch Institute for Medical Innovation, Radboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
6
|
Agoro R, Myslinski J, Marambio YG, Janosevic D, Jennings KN, Liu S, Hibbard LM, Fang F, Ni P, Noonan ML, Solis E, Chu X, Wang Y, Dagher PC, Liu Y, Wan J, Hato T, White KE. Dynamic single cell transcriptomics defines kidney FGF23/KL bioactivity and novel segment-specific inflammatory targets. Kidney Int 2025; 107:687-699. [PMID: 39828039 PMCID: PMC11928261 DOI: 10.1016/j.kint.2024.12.014] [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: 07/03/2023] [Revised: 11/22/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025]
Abstract
Fibroblast growth factor 23 (FGF23) via its coreceptor αKlotho (KL) provides critical control of phosphate metabolism, which is altered in both rare and very common syndromes. However, the spatial-temporal mechanisms dictating kidney FGF23 functions remain poorly understood. Thus, developing approaches to modify specific FGF23-dictated pathways has proven problematic. Herein, wild type mice were injected with rFGF23 for one, four and 12h and kidney FGF23 bioactivity was determined at single cell resolution. Computational analysis identified distinct epithelial, endothelial, stromal, and immune cell clusters, with differential expressional analysis uniquely tracking FGF23 bioactivity at each time point. FGF23 actions were sex independent but critically relied upon constitutive KL expression mapped within proximal tubule (segments S1-S3) and distal convoluted tub/connecting tubule cell sub-populations. Temporal KL-dependent FGF23 responses drove unique and transient cellular identities, including genes in key MAPK-signaling and vitamin D-metabolic pathways via early- (transcription factor AP-1-related) and late-phase (initiation factor EIF2 signaling) transcriptional regulons. Combining ATACseq/RNAseq data from a cell line stably expressing KL with the in vivo scRNAseq pinpointed genomic accessibility changes in MAPK-dependent genes, including the identification of FGF23-dependent early growth factor-1 distal enhancers. Finally, we identified unexpected crosstalk between FGF23-mediated MAPK signaling and pro inflammatory TNF receptor activation via transcription factor NF-κB, which blocked FGF23 bioactivity in vitro and in vivo. Collectively, our findings have uncovered novel pathways at the single cell level that likely influence FGF23-dependent disease mechanisms.
Collapse
Affiliation(s)
- Rafiou Agoro
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA; The Jackson Laboratory, Bar Harbor, Maine, USA.
| | - Jered Myslinski
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yamil G Marambio
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Danielle Janosevic
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kayleigh N Jennings
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Lainey M Hibbard
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Fang Fang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Pu Ni
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Megan L Noonan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Emmanuel Solis
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Xiaona Chu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yue Wang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Pierre C Dagher
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Takashi Hato
- The Jackson Laboratory, Bar Harbor, Maine, USA; Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, USA
| | - Kenneth E White
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
7
|
Xu Y, You J, Yao J, Hou B, Wang W, Hao Z. Klotho alleviates oxidative stress and mitochondrial dysfunction through the Nrf2/HO-1 pathway, thereby reducing renal senescence induced by calcium oxalate crystals. Urolithiasis 2025; 53:61. [PMID: 40156629 DOI: 10.1007/s00240-025-01734-z] [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: 01/03/2025] [Accepted: 03/15/2025] [Indexed: 04/01/2025]
Abstract
Klotho is an antiaging protein that is primarily secreted by the kidneys. This study aimed to explore the protective effects of Klotho against calcium oxalate (CaOx) crystal-induced renal aging and the underlying mechanisms involved. We established a mouse model of CaOx crystal deposition via the intraperitoneal injection of glyoxylate (Gly) and constructed an in vitro model by stimulating HK2 cells with calcium oxalate monohydrate (COM). Renal aging levels were assessed through β-galactosidase (SA-β-gal) staining and the detection of senescence-associated markers. By overexpressing Klotho both in vitro and in vivo, we examined oxidative stress, mitochondrial function, and renal aging levels. We then evaluated the role of Nrf2/HO-1 signalling pathway-mediated oxidative stress in CaOx crystal-induced renal aging by applying the oxidative stress scavenger N-acetylcysteine (NAC) and overexpressing or inhibiting Nrf2 in HK2 cells. We subsequently overexpressed Klotho while inhibiting Nrf2 to confirm that Klotho exerts its protective effects through the Nrf2/HO-1 pathway. Finally, we measured the methylation levels of the Klotho promoter and assessed the degree of renal aging induced by CaOx crystals after the inhibition of Klotho DNA methylation. We found that the overexpression of Klotho alleviated CaOx crystal-induced oxidative stress and mitochondrial dysfunction, thereby reducing renal aging. NAC mitigated CaOx crystal-induced renal aging. The overexpression of Nrf2 alleviated CaOx crystal-induced oxidative stress and mitochondrial dysfunction, thus reducing renal aging, whereas the knockdown of Nrf2 exacerbated CaOx crystal-induced oxidative stress and mitochondrial dysfunction, leading to more severe renal aging. The combination of Klotho overexpression and Nrf2 knockdown reversed the protective effects of Klotho. CaOx crystals induced an increase in the DNA methylation levels of Klotho in the kidneys, and the inhibition of DNA methylation alleviated CaOx-induced renal aging. This study revealed that Klotho plays a crucial role in calcium oxalate crystal-induced kidney senescence by influencing kidney oxidative stress and mitochondrial function through the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Yuexian Xu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China
| | - Jianmin You
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China
| | - Junfeng Yao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China
| | - Bingbing Hou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
- Institute of Urology, Anhui Medical University, Hefei, China.
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China.
| | - Wei Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
- Institute of Urology, Anhui Medical University, Hefei, China.
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China.
- Department of Urology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China.
| | - Zongyao Hao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
- Institute of Urology, Anhui Medical University, Hefei, China.
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China.
| |
Collapse
|
8
|
Wagner CA, Frey-Wagner I, Ortiz A, Unwin R, Liabeuf S, Suzumoto Y, Iervolino A, Stasi A, Di Marzo V, Gesualdo L, Massy ZA. The role of the intestinal microbiome in cognitive decline in patients with kidney disease. Nephrol Dial Transplant 2025; 40:ii4-ii17. [PMID: 40080091 PMCID: PMC11905753 DOI: 10.1093/ndt/gfae253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Indexed: 03/15/2025] Open
Abstract
Cognitive decline is frequently seen in patients with chronic kidney disease (CKD). The causes of cognitive decline in these patients are likely to be multifactorial, including vascular disease, uraemic toxins, blood-brain barrier leakage, and metabolic and endocrine changes. Gut dysbiosis is common in patients with CKD and contributes to the increase in uraemic toxins. However, the gut microbiome modulates local and systemic levels of several metabolites such as short-chain fatty acids or derivatives of tryptophan metabolism, neurotransmitters, endocannabinoid-like mediators, bile acids, hormones such as glucagon-like peptide 1 (GLP1) or cholecystokinin (CCK). These factors can affect gut function, immunity, autonomic nervous system activity and various aspects of brain function. Key areas include blood-brain barrier integrity, nerve myelination and survival/proliferation, appetite, metabolism and thermoregulation, mood, anxiety and depression, stress and local inflammation. Alterations in the composition of the gut microbiota and the production of biologically active metabolites in patients with CKD are well documented and are favoured by low-fiber diets, elevated urea levels, sedentary lifestyles, slow stool transit times and polypharmacy. In turn, dysbiosis can modulate brain function and cognitive processes, as discussed in this review. Thus, the gut microbiome may contribute to alterations in cognition in patients with CKD and may be a target for therapeutic interventions using diet, prebiotics and probiotics.
Collapse
Affiliation(s)
- Carsten A Wagner
- Institute of Physiology and Zurich Kidney Center, University of Zurich, Switzerland
| | | | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, RICORS2040, Madrid, Spain
| | - Robert Unwin
- Department of Renal Medicine, University College London, London, UK
| | - Sophie Liabeuf
- Pharmacoepidemiology Unit, Department of Clinical Pharmacology, Amiens-Picardie University Medical Center, Amiens, France
- MP3CV Laboratory, Jules Verne University of Picardie, Amiens, France
| | - Yoko Suzumoto
- Biogem, Molecular Biology and Genetics Research Institute, Ariano Irpino, Italy
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Naples, Italy
| | - Anna Iervolino
- Biogem, Molecular Biology and Genetics Research Institute, Ariano Irpino, Italy
- University of Campania “L. Vanvitelli”, Naples, Italy
| | - Alessandra Stasi
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J) Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Di Marzo
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, CRIUCPQ and INAF, Centre NUTRISS, Faculties of Medicine and Agriculture and Food Sciences, Université Laval, Québec City, Canada
- Joint International Research Unit for Chemical and Biomolecular Research on the Microbiome and its impact on Metabolic Health and Nutrition (JIRU-MicroMeNu) between Université Laval Québec, Canada and Consiglio Nazionale delle Ricerche, Institute of Biomolecular Chemistry, Pozzuoli, Italy
| | - Loreto Gesualdo
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J) Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Ziad A Massy
- INSERM Unit 1018, Team 5, CESP, Hôpital Paul Brousse, Paris-Saclay University and Versailles Saint-Quentin-en-Yvelines University (UVSQ), Villejuif, France
- Association pour l'Utilisation du Rein Artificiel dans la région parisienne (AURA) Paris, France and Ambroise Paré University Hospital, APHP, Department of Nephrology Boulogne-Billancourt, Paris, France
| |
Collapse
|
9
|
Gerber JS, Arroyo EMP, Pastor J, Correia M, Rudloff S, Moe OW, Egli-Spichtig D, Mohebbi N, Wagner CA. Controlled dietary phosphate loading in healthy young men elevates plasma phosphate and FGF23 levels. Pflugers Arch 2025; 477:495-508. [PMID: 39601886 PMCID: PMC11825603 DOI: 10.1007/s00424-024-03046-4] [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: 07/16/2024] [Revised: 11/01/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Increased dietary inorganic phosphate (Pi) intake stimulates renal Pi excretion, in part, by parathyroid hormone (PTH), fibroblast growth factor 23 (FGF23) or dopamine. High dietary Pi may also stimulate sympathetic outflow. Rodent studies provided evidence for these regulatory loops, while controlled experiments in healthy humans examined periods of either a few hours or several weeks, and often varied dietary calcium intake. The effects of controlled, isolated changes in dietary Pi intake over shorter periods are unknown. We studied the effects of a low or high Pi diet on parameters of mineral metabolism in 10 healthy young men. Participants received a standardized diet (1000 mg phosphorus equivalent/day) supplemented with either a phosphate binder (low Pi diet) or phosphate capsules (750 mg phosphorus, high Pi diet) in a randomized cross-over trial for 5 days with a 7-day washout between diets. High Pi intake increased plasma Pi levels and 24-h excretion and decreased urinary calcium excretion. High Pi intake increased intact FGF23 (iFGF23) and suppressed plasma Klotho without affecting cFGF23, PTH, calcidiol, calcitriol, Fetuin-A, dopamine, epinephrine, norepinephrine, metanephrine, or aldosterone. Higher iFGF23 correlated with lower calcitriol and higher PTH. These data support a role for iFGF23 in increasing renal Pi excretion and reducing calcitriol in healthy young men during steady-state high dietary Pi intake. High dietary Pi intake elevated blood Pi levels in healthy young subjects with normal renal function and may therefore be a health risk, as higher serum Pi levels are associated with cardiovascular risk in the general population.
Collapse
Affiliation(s)
- Jennifer Scotti Gerber
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Eva Maria Pastor Arroyo
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Johanne Pastor
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Miguel Correia
- Division of Nephrology and Hypertension, University of Bern and University Hospital Bern, Bern, Switzerland
| | - Stefan Rudloff
- Division of Nephrology and Hypertension, University of Bern and University Hospital Bern, Bern, Switzerland
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Daniela Egli-Spichtig
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
- National Center of Competence in Research, NCCR Kidney.CH, Zurich, Switzerland
| | - Nilufar Mohebbi
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
- National Center of Competence in Research, NCCR Kidney.CH, Zurich, Switzerland
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
- National Center of Competence in Research, NCCR Kidney.CH, Zurich, Switzerland.
| |
Collapse
|
10
|
Wen Z, Liu X, Zhang T, Zhang J, Zou Z, Jiang S, Li S. Association between weight-adjusted-waist index and serum anti-aging protein α-Klotho in U.S. adults: Evidence from NHANES 2007-2016. Exp Gerontol 2025; 200:112672. [PMID: 39765322 DOI: 10.1016/j.exger.2024.112672] [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: 09/14/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND As unhealthy diets have proliferated, there has been an increasing trend in the prevalence of obesity along with socioeconomic progress. WWI, mainly representing weight-independent central obesity, is a more accurate indicator of obesity than BMI and WC. Klotho is known to be one of the root causes of several age-related illnesses. There has never been any prior evidence associating WWI and serum α-Klotho levels. METHODS The current cross-sectional investigation used data from the National Health and Nutrition Examination Survey (NHANES) 2013-2016. The calculation of each participant's WWI involved dividing their waist circumference in centimeters by the square root of their weight in kilograms. Using multivariable linear regression models, the association between serum Klotho concentrations and WWI was analyzed. Smoothed curve fitting was utilized to investigate the nonlinear connection. In addition, interaction tests and other subgroup analyses were carried out. RESULTS With an average age of 57.57 ± 10.81 years, 13,172 participants were enrolled in this study, of whom 51.45 % were female. After full adjustment, each unit increase in WWI was significantly associated with a decrease in Klotho by 23.18 ng/ml [β(95%CI) = -23.18 (-31.34, -15.02)]. Moreover, we uncovered that WWI had a stronger negative correlation with Klotho than other obesity markers, such as WC, body mass index (BMI), and a body shape index (ABSI). CONCLUSIONS To sum up, weight-adjusted-waist index levels were negatively associated with serum Klotho protein concentration and showed a stronger correlation than other obesity markers. Our findings indicated that WWI may be a predictor of reduced Klotho levels, and that controlling obesity in accordance with WWI may help lower the Klotho levels.
Collapse
Affiliation(s)
- Zujun Wen
- Department of pharmacy, Heyuan People's Hospital, Heyuan, China.
| | - Xiang Liu
- The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Tingting Zhang
- Department of pharmacy, Heyuan People's Hospital, Heyuan, China
| | - Junhai Zhang
- Department of burns and plastic surgery, Heyuan People's Hospital, Heyuan, China
| | - Zhicong Zou
- Department of pharmacy, Heyuan People's Hospital, Heyuan, China
| | - Shuncheng Jiang
- Department of pharmacy, Heyuan People's Hospital, Heyuan, China
| | - Shusi Li
- Department of pharmacy, Heyuan People's Hospital, Heyuan, China
| |
Collapse
|
11
|
Ortega MA, Boaru DL, De Leon-Oliva D, De Castro-Martinez P, Minaya-Bravo AM, Casanova-Martín C, Barrena-Blázquez S, Garcia-Montero C, Fraile-Martinez O, Lopez-Gonzalez L, Saez MA, Alvarez-Mon M, Diaz-Pedrero R. The Impact of Klotho in Cancer: From Development and Progression to Therapeutic Potential. Genes (Basel) 2025; 16:128. [PMID: 40004457 PMCID: PMC11854833 DOI: 10.3390/genes16020128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Klotho, initially identified as an anti-aging gene, has been shown to play significant roles in cancer biology. Alongside α-Klotho, the β-Klotho and γ-Klotho isoforms have also been studied; these studies showed that Klotho functions as a potential tumor suppressor in many different cancers by inhibiting cancer cell proliferation, inducing apoptosis and modulating critical signaling pathways such as the Wnt/β-catenin and PI3K/Akt pathways. In cancers such as breast cancer, colorectal cancer, hepatocellular carcinoma, ovarian cancer, and renal cell carcinoma, reduced Klotho expression often correlates with a poor prognosis. In addition, Klotho's role in enhancing chemotherapy sensitivity and its epigenetic regulation further underscores its potential as a target for cancer treatments. This review details Klotho's multifaceted contributions to cancer suppression and its potential as a therapeutic target, enhancing the understanding of its significance in cancer treatment and prognoses.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Ana M. Minaya-Bravo
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Carlos Casanova-Martín
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Silvestra Barrena-Blázquez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Department of General and Digestive Surgery, Príncipe de Asturias, University Hospital, 28805 Alcala de Henares, Spain
| | - Cielo Garcia-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Miguel A. Saez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Pathological Anatomy Service, Central University Hospital of Defence—UAH Madrid, 28801 Alcala de Henares, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, 28806 Alcala de Henares, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Department of General and Digestive Surgery, Príncipe de Asturias, University Hospital, 28805 Alcala de Henares, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| |
Collapse
|
12
|
Rivoira MA, Peralta López ME, Areco V, Díaz de Barboza G, Dionisi MP, Tolosa de Talamoni N. Emerging concepts on the FGF23 regulation and activity. Mol Cell Biochem 2025; 480:75-89. [PMID: 38581553 DOI: 10.1007/s11010-024-04982-6] [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: 12/20/2023] [Accepted: 02/28/2024] [Indexed: 04/08/2024]
Abstract
Fibroblast growth factor 23 (FGF23) discovery has provided new insights into the regulation of Pi and Ca homeostasis. It is secreted by osteoblasts and osteocytes, and acts mainly in the kidney, parathyroid, heart, and bone. The aim of this review is to highlight the current knowledge on the factors modulating the synthesis of FGF23, the canonical and non-canonical signaling pathways of the hormone, the role of FGF23 in different pathophysiological conditions, and the anti-FGF23 therapy. This is a narrative review based on the search of PubMed database in the range of years 2000-2023 using the keywords local and systemic regulators of FGF23 synthesis, FGF23 receptors, canonical and non-canonical pathways, pathophysiological conditions and FGF23, and anti-FGF23 therapy, focusing the data on the molecular mechanisms. The regulation of FGF23 synthesis is complex and multifactorial. It is regulated by local factors and systemic regulators mainly involved in bone mineralization. The excessive FGF23 production is associated with different congenital diseases and with diseases occurring with a secondary high FGF23 production such as in chronic disease kidney and tumor-induced osteomalacia (TIO). The anti-FGF23 therapy appears to be useful to treat chromosome X-linked hypophosphatemia and TIO, but there are doubts about the handle of excessive FGF23 production in CKD. FGF23 biochemistry and pathophysiology are generating a plethora of knowledge to reduce FGF23 bioactivity at many levels that might be useful for future therapeutics of diseases associated with high-serum FGF23 levels.
Collapse
Affiliation(s)
- María Angélica Rivoira
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do, Piso, Ciudad Universitaria, 5000, Córdoba, Argentina
| | - María Elena Peralta López
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do, Piso, Ciudad Universitaria, 5000, Córdoba, Argentina
| | - Vanessa Areco
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do, Piso, Ciudad Universitaria, 5000, Córdoba, Argentina
- Instituto Multidisciplinario de Investigación y Transferencia Agroalimentaria y Biotecnológica (IMITAB, CONICET-UNVM), Córdoba, Argentina
| | - Gabriela Díaz de Barboza
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do, Piso, Ciudad Universitaria, 5000, Córdoba, Argentina
| | - María Paula Dionisi
- Cátedra de Clínica Médica II - UHMI Nº 2, Hospital San Roque, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nori Tolosa de Talamoni
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do, Piso, Ciudad Universitaria, 5000, Córdoba, Argentina.
| |
Collapse
|
13
|
Xu P, Jiang M, Chen J, Zhou Y, Wang Z. The Long-Range Chromosomal Interaction Controlling Klotho Gene Expression in Human Chronic Kidney Disease. ACS OMEGA 2024; 9:51264-51270. [PMID: 39758635 PMCID: PMC11696417 DOI: 10.1021/acsomega.4c07967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/22/2024] [Accepted: 12/02/2024] [Indexed: 01/07/2025]
Abstract
Cis-regulatory elements bridge enhancers and gene promoters to control gene expression via distal DNA interaction and three-dimensional chromosomal conformation organization. The aberrant changes of cis-acting regulatory systems as one type of the epigenetic regulative ways may be connected with human genetic diseases. Klotho, as an antiaging protein, is selectively expressed in kidney tissues and plays a crucial role in preventing chronic kidney disease (CKD) and renal fibrosis. However, the underlying transcription regulatory mechanism of Klotho in CKD is not fully understood. Herein, we analyzed the spatial organization of the chromatin region spanning 2 Mb upstream Klotho in human renal punctured CKD tissues using chromosome conformation capture (3C)-qPCR and identified the distal interaction of the Klotho promoter with certain specific chromatin regions characterized as the regulatory elements. Moreover, we determined that four DNase I hypersensitive sites (DHSs) involved in the regulation of Klotho gene expression lost their activities in CKD tissues compared to control accompanied by the reduction of H3K27ac. Finally, the CCCTC-binding factor (CTCF) sites were validated on the DHSs beyond the Klotho promoter by chromatin looping formation through the recruitment of CTCF.
Collapse
Affiliation(s)
- Pengwei Xu
- Department of Urology, Suzhou Ninth Hospital affiliated to Soochow University, Suzhou 215000, China
| | - Minjun Jiang
- Department of Urology, Suzhou Ninth Hospital affiliated to Soochow University, Suzhou 215000, China
| | - Jianchun Chen
- Department of Urology, Suzhou Ninth Hospital affiliated to Soochow University, Suzhou 215000, China
| | - Yongqiang Zhou
- Department of Urology, Suzhou Ninth Hospital affiliated to Soochow University, Suzhou 215000, China
| | - Zhenfan Wang
- Department of Urology, Suzhou Ninth Hospital affiliated to Soochow University, Suzhou 215000, China
| |
Collapse
|
14
|
Konnur A, Gang S, Hegde U, Patel H, Pandya A, Shete N. Chronic Kidney Disease: Decreasing Serum Klotho Levels Predict Adverse Renal and Vascular Outcomes. Int J Nephrol 2024; 2024:2803739. [PMID: 39544340 PMCID: PMC11563715 DOI: 10.1155/2024/2803739] [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: 07/02/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
Background and Objectives: Soluble alpha Klotho (s.Klotho) is an emerging marker for chronic kidney disease (CKD) prognosis. The objective was to study the association between s.Klotho and CKD-related decrease in glomerular filtration rate (GFR), bone and vascular damage. Method: A total of 118 patients with CKD stage 2-4 were enrolled and 107 patients continued in the study. Clinical and laboratory parameters were recorded at time of enrollment and 12 months. A double sandwich ELISA for s.Klotho was recorded in controls (n = 25) and patients' serum samples at 6 months (n = 107) and 12 months (n = 102). Primary endpoints like 40% or more fall in GFR, a requirement for renal replacement therapy (RRT), and death with different grades of s.Klotho deficiency were studied. Results: Of the 107 patients (80 male and 27 female), mean s.Klotho was 3.46 ng/mL (02.3-04.2). The GFR fall was significantly different (p value < 0.0001) in the different grades of s.Klotho deficiency with Grade 4 s.Klotho deficiency (0.1-2.99 ng/mL) having the maximum fall of GFR at 9.2 mL/min/1.73 m2 (04.8-12.0) and minimum in Grade 2 (3-5.99 ng/mL) at 1.35 mL/min/1.73 m2 (03.0-02.75). The Ankle Brachial Pressure Index positively correlated with s.Klotho and the correlation coefficient was 0.536 (0.382-0.662) (p < 0.001). The carotid intimal medial thickness negatively correlated with s.Klotho and the correlation coefficient was -0.712 (95% CI: -0.797--0.601, p < 0.001). All five deaths had s.Klotho Grade 4 (severe) deficiency. The event-free survival rate was maximum (100%) in Grade 2 Klotho deficiency and lowest (55%) in Grade 4 s.Klotho deficiency. Conclusions: s.Klotho levels decreased significantly in patients with progressive kidney failure. s.Klotho levels significantly correlated with the presence of vascular disease. Death and need for RRT were significantly more in patients with severe s.Klotho deficiency.
Collapse
Affiliation(s)
- Abhijit Konnur
- Department of Nephrology, Muljibhai Patel Urological Hospital, Nadiad, India
| | - Sishir Gang
- Department of Nephrology, Muljibhai Patel Urological Hospital, Nadiad, India
| | - Umapati Hegde
- Department of Nephrology, Muljibhai Patel Urological Hospital, Nadiad, India
| | - Hardik Patel
- Department of Nephrology, Muljibhai Patel Urological Hospital, Nadiad, India
| | - Akash Pandya
- Department of Nephrology, Muljibhai Patel Urological Hospital, Nadiad, India
| | - Nitiraj Shete
- Department of Biostatistics, Muljibhai Patel Urological Hospital, Nadiad, India
| |
Collapse
|
15
|
Guo Y, Wan F, Shi YP, Zhang HM, Yang RC. Association between renal α-klotho and renal pathology among patients with chronic kidney disease. Ther Apher Dial 2024; 28:769-774. [PMID: 38818966 DOI: 10.1111/1744-9987.14165] [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: 02/26/2024] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 06/01/2024]
Abstract
INTRODUCTION This study was designed to investigate whether renal α-klotho levels are associated with renal pathology. This is the first report on patients with chronic kidney disease (CKD). METHODS We conducted a retrospective observational study. A total of 65 CKD patients were enrolled. Serum and renal biopsy samples were collected. Estimated glomerular filtration rate (eGFR) was examined by biochemical test. And α-klotho expressions were assessed by RT-PCR and immunohistochemistry. In addition, detailed microscopic findings were reviewed. RESULTS Renal α-klotho levels are associated positively with eGFR, and negatively with renal pathology, including interstitial fibrosis, inflammatory cell infiltration, and tubular atrophy. CONCLUSIONS The renal α-klotho is related to renal pathology.
Collapse
Affiliation(s)
- Yan Guo
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, China
| | - Feng Wan
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, China
| | - Yan-Peng Shi
- Linping Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, China
| | - Hong-Mei Zhang
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ru-Chun Yang
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, China
| |
Collapse
|
16
|
Grigore TV, Zuidscherwoude M, Olauson H, Hoenderop JG. Lessons from Klotho mouse models to understand mineral homeostasis. Acta Physiol (Oxf) 2024; 240:e14220. [PMID: 39176993 DOI: 10.1111/apha.14220] [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: 05/31/2024] [Revised: 07/30/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024]
Abstract
AIM Klotho, a key component of the endocrine fibroblast growth factor receptor-fibroblast growth factor axis, is a multi-functional protein that impacts renal electrolyte handling. The physiological significance of Klotho will be highlighted in the regulation of calcium, phosphate, and potassium metabolism. METHODS In this review, we compare several murine models with different renal targeted deletions of Klotho and the insights into the molecular and physiological function that these models offer. RESULTS In vivo, Klotho deficiency is associated with severely impaired mineral metabolism, with consequences on growth, longevity and disease development. Additionally, we explore the perspectives of Klotho in renal pathology and vascular events, as well as potential Klotho treatment options. CONCLUSION This comprehensive review emphasizes the use of Klotho to shed light on deciphering the renal molecular in vivo mechanisms in electrolyte handling, as well as novel therapeutic interventions.
Collapse
Affiliation(s)
- Teodora V Grigore
- Department of Medical BioSciences, Radboud Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Malou Zuidscherwoude
- Department of Medical BioSciences, Radboud Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hannes Olauson
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Joost G Hoenderop
- Department of Medical BioSciences, Radboud Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
17
|
Yamada H, Kuro-O M, Funazaki S, Hamamoto K, Hara K. Soluble αKlotho concentration in the inferior vena cava of patients with primary aldosteronism. Nefrologia 2024; 44:623-627. [PMID: 39547775 DOI: 10.1016/j.nefroe.2024.11.003] [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: 12/22/2023] [Accepted: 04/21/2024] [Indexed: 11/17/2024] Open
Abstract
INTRODUCTION Klotho, a key aging regulator, is predominantly expressed in the kidney. Various methods now enable the measurement of soluble αKlotho blood levels in humans. Limited studies have explored the renal origin of circulating αKlotho in humans. METHODS Soluble αKlotho in the inferior vena cava blood was measured using an enzyme-linked immunosorbent assay kit using blood samples from patients undergoing adrenal venous catheterization for close examination of primary aldosteronism. RESULTS The concentration at the suprarenal inferior vena cava (476±68.2) was significantly higher than that at the infrarenal inferior vena cava (434±74.8) (p=0.018), with a rate of change of 8.12 (2.3)%. CONCLUSIONS We demonstrate a step-up in αKlotho concentration from the infrarenal to suprarenal vena cava in humans, supporting the kidney's origin of soluble αKlotho in the bloodstream.
Collapse
Affiliation(s)
- Hodaka Yamada
- Department of Medicine, Division of Endocrinology and Metabolism, Jichi Medical University Saitama Medical Center, 1-847 Amanuma-cho, Omiya-ku, Saitama 330-8503, Japan.
| | - Makoto Kuro-O
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1, Shimotsuke, Tochigi 329-0498, Japan
| | - Shunsuke Funazaki
- Department of Medicine, Division of Endocrinology and Metabolism, Jichi Medical University Saitama Medical Center, 1-847 Amanuma-cho, Omiya-ku, Saitama 330-8503, Japan
| | - Kohei Hamamoto
- Department of Radiology, School of Medicine, Jichi Medical University Saitama Medical Center, 1-847 Amanuma-cho, Omiya-ku, Saitama 330-8503, Japan
| | - Kazuo Hara
- Department of Medicine, Division of Endocrinology and Metabolism, Jichi Medical University Saitama Medical Center, 1-847 Amanuma-cho, Omiya-ku, Saitama 330-8503, Japan
| |
Collapse
|
18
|
Tang Y, Jiang J, Zhao Y, Du D. Aging and chronic kidney disease: epidemiology, therapy, management and the role of immunity. Clin Kidney J 2024; 17:sfae235. [PMID: 40034487 PMCID: PMC11873799 DOI: 10.1093/ckj/sfae235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Indexed: 03/05/2025] Open
Abstract
Chronic kidney disease (CKD) is now an unquestionable progressive condition that affects more than 10% of the general population worldwide, and has emerged as one of the most important causes of global mortality. It is clear that the prevalence of CKD among the aging population is significantly elevated. It involves a broad range of complex and poorly understood concerns in older adults such as frailty, malnutrition, sarcopenia, and even cognitive and mental dysfunction. In kidneys, renal function such as glomerular filtration, urine concentration and dilution, and homeostasis of sodium and potassium, can be influenced by the aging process. In addition, it is worth noting that CKD and end-stage kidney disease patients often have accompanying activation of immune system and inflammation, involving both the innate and adaptive immune system. Based on this background, in this review article we attempt to summarize the epidemiological characteristics of CKD in the aging population, discuss the immunological mechanisms in aging-related CKD, and furnish the reader with processes for the therapy and management of elderly patients with CKD.
Collapse
Affiliation(s)
- Yukun Tang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences; Wuhan, China
| | - Jipin Jiang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences; Wuhan, China
| | - Yuanyuan Zhao
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences; Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
| | - Dunfeng Du
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences; Wuhan, China
| |
Collapse
|
19
|
Huang X, Hu L, Tao S, Xue T, Hou C, Li J. Relationship between uric acid to high-density cholesterol ratio (UHR) and circulating α-klotho: evidence from NHANES 2007-2016. Lipids Health Dis 2024; 23:244. [PMID: 39123222 PMCID: PMC11312937 DOI: 10.1186/s12944-024-02234-6] [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: 06/08/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
OBJECTIVE To investigate the relationship between uric acid to high-density lipoprotein cholesterol ratio (UHR) and circulating α-klotho levels in U.S. adults. METHODS A cross-sectional study used data from the National Health and Nutrition Examination Survey (NHANES) from 2007 to 2016. Circulating α-klotho was defined as the dependent variable and UHR was defined as the independent variable. Multivariable linear regression was performed to assess the relationship between the independent and dependent variables. The nonlinear relationship and effect size between UHR and α-klotho were evaluated using smooth curve fitting and threshold effect analysis. Subgroup analysis and sensitivity analysis were conducted to determine the stability of the results. The diagnostic performance of UHR and α-klotho in common elderly diseases was compared using ROC (Receiver Operating Characteristic) analysis. RESULTS Among 12,849 participants, there was a negative relationship between the UHR and circulating α-klotho. In the fully adjusted overall model, each unit increase in UHR was associated with a decrease of 4.1 pg/mL in α-klotho. The threshold effect analysis showed that before the inflection point of 8.2, each unit increase in UHR was associated with a decrease of 15.0 pg/mL in α-klotho; beyond the inflection point of 8.2, each unit increase in UHR was associated with a decrease of 2.8 pg/mL in α-klotho. Subgroup analyses and sensitivity analysis indicated that the relationship between UHR and α-klotho remained stable across most populations. The ROC diagnostic test indicated that the evaluative efficacy of UHR in diagnosing age-related diseases was comparable to that of α-klotho. CONCLUSION This study revealed that the UHR was associated with the circulating α-klotho concentration, with a negative association observed in most cases. This finding suggested that the UHR might be a promising indicator for evaluating circulating α-klotho levels.
Collapse
Affiliation(s)
- Xuanchun Huang
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Lanshuo Hu
- Xiyuan Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Shiyi Tao
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Tiantian Xue
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Chengzhi Hou
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China.
| | - Jun Li
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China.
| |
Collapse
|
20
|
Marečáková N, Kačírová J, Tóthová C, Maďari A, Maďar M, Farbáková J, Horňák S. Determination of the reference interval for urinary klotho to creatinine ratio of healthy dogs. Front Vet Sci 2024; 11:1423390. [PMID: 39113723 PMCID: PMC11305118 DOI: 10.3389/fvets.2024.1423390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
For several years, alpha klotho has been considered as a candidate biomarker in chronic kidney disease (CKD), progression of CKD and CKD mineral bone disorders (CKD-MBD). The evidence on the relationship between klotho and kidney function is controversial in some areas. The aim of the study was to identify the influence of age, sex and breed on urinary alpha klotho, values in the early stages of CKD within the studied population and determine a reference interval in a group of healthy dogs. Significantly higher values were measured in older dogs over 6 years old (p = 0.026, p = 0.0007) and in the breed German Shepherd than Belgian Shepherd (p = 0.0401). On the basis of sex and in small breed dogs, no significant differences were noted. In dogs with CKD stage 2, alpha klotho values were significantly lower (p = 0.0135) than in healthy dogs. Within the studied population, a reference interval for urinary klotho to creatinine ratio (UrKl/Cr) was determined in the range of 3.94-23.55 pg/gCr. Since our findings show that alpha klotho is associated with older age, we assume that this may have influenced the results in the group of dogs with CKD stage 1 due to the presence of predominantly old dogs in this group. Future studies would be needed to consider age as a factor affecting urinary alpha klotho in dogs with CKD.
Collapse
Affiliation(s)
- Nikola Marečáková
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovakia
| | - Jana Kačírová
- Institute of Plant Genetics and Biotechnology, Plant Science and Biodiversity Centre, Slovak Academy of Sciences, Nitra, Slovakia
| | - Csilla Tóthová
- Clinic of Ruminants, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovakia
| | - Aladár Maďari
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovakia
| | - Marián Maďar
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovakia
| | - Jana Farbáková
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovakia
| | - Slavomír Horňák
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovakia
| |
Collapse
|
21
|
Agoro R, Myslinski J, Marambio YG, Janosevic D, Jennings KN, Liu S, Hibbard LM, Fang F, Ni P, Noonan ML, Solis E, Chu X, Wang Y, Dagher PC, Liu Y, Wan J, Hato T, White KE. Dynamic Single Cell Transcriptomics Defines Kidney FGF23/KL Bioactivity and Novel Segment-Specific Inflammatory Targets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595014. [PMID: 38853876 PMCID: PMC11160572 DOI: 10.1101/2024.05.24.595014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
FGF23 via its coreceptor αKlotho (KL) provides critical control of phosphate metabolism, which is altered in rare and very common syndromes, however the spatial-temporal mechanisms dictating renal FGF23 functions remain poorly understood. Thus, developing approaches to modify specific FGF23-dictated pathways has proven problematic. Herein, wild type mice were injected with rFGF23 for 1, 4 and 12h and renal FGF23 bioactivity was determined at single cell resolution. Computational analysis identified distinct epithelial, endothelial, stromal, and immune cell clusters, with differential expressional analysis uniquely tracking FGF23 bioactivity at each time point. FGF23 actions were sex independent but critically relied upon constitutive KL expression mapped within proximal tubule (S1-S3) and distal tubule (DCT/CNT) cell sub-populations. Temporal KL-dependent FGF23 responses drove unique and transient cellular identities, including genes in key MAPK- and vitamin D-metabolic pathways via early- (AP-1-related) and late-phase (EIF2 signaling) transcriptional regulons. Combining ATACseq/RNAseq data from a cell line stably expressing KL with the in vivo scRNAseq pinpointed genomic accessibility changes in MAPK-dependent genes, including the identification of FGF23-dependent EGR1 distal enhancers. Finally, we isolated unexpected crosstalk between FGF23-mediated MAPK signaling and pro-inflammatory TNF receptor activation via NF-κB, which blocked FGF23 bioactivity in vitro and in vivo . Collectively, our findings have uncovered novel pathways at the single cell level that likely influence FGF23-dependent disease mechanisms. Translational statement Inflammation and elevated FGF23 in chronic kidney disease (CKD) are both associated with poor patient outcomes and mortality. However, the links between these manifestations and the effects of inflammation on FGF23-mediated mineral metabolism within specific nephron segments remain unclear. Herein, we isolated an inflammatory pathway driven by TNF/NF-κB associated with regulating FGF23 bioactivity. The findings from this study could be important in designing future therapeutic approaches for chronic mineral diseases, including potential combination therapies or early intervention strategies. We also suggest that further studies could explore these pathways at the single cell level in CKD models, as well as test translation of our findings to interactions of chronic inflammation and elevated FGF23 in human CKD kidney datasets.
Collapse
|
22
|
Jung HJ, Pham TD, Su XT, Grigore TV, Hoenderop JG, Olauson H, Wall SM, Ellison DH, Welling PA, Al-Qusairi L. Klotho is highly expressed in the chief sites of regulated potassium secretion, and it is stimulated by potassium intake. Sci Rep 2024; 14:10740. [PMID: 38729987 PMCID: PMC11087591 DOI: 10.1038/s41598-024-61481-w] [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: 10/27/2023] [Accepted: 05/06/2024] [Indexed: 05/12/2024] Open
Abstract
Klotho regulates many pathways in the aging process, but it remains unclear how it is physiologically regulated. Because Klotho is synthesized, cleaved, and released from the kidney; activates the chief urinary K+ secretion channel (ROMK) and stimulates urinary K+ secretion, we explored if Klotho protein is regulated by dietary K+ and the potassium-regulatory hormone, Aldosterone. Klotho protein along the nephron was evaluated in humans and in wild-type (WT) mice; and in mice lacking components of Aldosterone signaling, including the Aldosterone-Synthase KO (AS-KO) and the Mineralocorticoid-Receptor KO (MR-KO) mice. We found the specific cells of the distal nephron in humans and mice that are chief sites of regulated K+ secretion have the highest Klotho protein expression along the nephron. WT mice fed K+-rich diets increased Klotho expression in these cells. AS-KO mice exhibit normal Klotho under basal conditions but could not upregulate Klotho in response to high-K+ intake in the K+-secreting cells. Similarly, MR-KO mice exhibit decreased Klotho protein expression. Together, i) Klotho is highly expressed in the key sites of regulated K+ secretion in humans and mice, ii) In mice, K+-rich diets increase Klotho expression specifically in the potassium secretory cells of the distal nephron, iii) Aldosterone signaling is required for Klotho response to high K+ intake.
Collapse
Affiliation(s)
- Hyun Jun Jung
- Department of Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Truyen D Pham
- Department of Nephrology, Emory University School of Medicine, Atlanta, GA, USA
| | - Xiao-Tong Su
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, USA
| | - Teodora Veronica Grigore
- Department of Medical BioSciences, Radboud Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost G Hoenderop
- Department of Medical BioSciences, Radboud Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hannes Olauson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Susan M Wall
- Department of Nephrology, Emory University School of Medicine, Atlanta, GA, USA
| | - David H Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, USA
| | - Paul A Welling
- Department of Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lama Al-Qusairi
- Department of Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
23
|
Koike M, Sato T, Shiozaki Y, Komiya A, Miura M, Higashi A, Ishikawa A, Takayanagi K, Uga M, Miyamoto KI, Segawa H. Involvement of α-klotho in growth hormone (GH) signaling. J Clin Biochem Nutr 2024; 74:221-229. [PMID: 38799134 PMCID: PMC11111466 DOI: 10.3164/jcbn.23-127] [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: 12/24/2023] [Accepted: 01/10/2024] [Indexed: 05/29/2024] Open
Abstract
Growth hormone (GH) exerts multiple effects on different organs directly or via its main mediator, insulin-like growth factor1 (IGF1). In this study, we focused on the novel relationship between GH action and the antiaging hormone α-klotho. Immunofluorescent staining of α-klotho was observed in the renal distal tubules and pituitary glands of somatostatin- and GH-positive cells in wild-type (WT) mice. Treatment of 4-week-old WT mice with GH increased IGF1 mRNA expression in the pituitary gland, liver, heart, kidney, and bone but increased α-klotho mRNA expression only in the pituitary gland, kidney, and bone. Increased α-klotho protein levels were observed in the kidney but not in the pituitary gland. No induction of α-klotho RNA expression by GH was observed in juvenile mice with kidney disease, indicating GH resistance. Furthermore, GH and α-klotho supplementation in HEK293 cells transfected with GHR increased Janus kinase 2 mRNA (a GH downstream signal) expression compared to supplementation with GH alone. In conclusion, we suggest that 1) the kidney is the main source of secreted α-klotho, which is detected in blood by the downstream action of GH, 2) α-klotho induction by GH is resistant in kidney disease, and 3) α-klotho might be an enhanced regulator of GH signaling.
Collapse
Affiliation(s)
- Megumi Koike
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Tetsuhiko Sato
- General Medicine, Nagoya Daini Red Cross Hospital, 2-9 Myoken-cho, Showa-ku, Nagoya 466-8650, Japan
| | - Yuji Shiozaki
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Aoi Komiya
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Mizuki Miura
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Ayami Higashi
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Akane Ishikawa
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Kaori Takayanagi
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Minori Uga
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Ken-ichi Miyamoto
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
- Graduate School of Agriculture, Ryukoku University, 1-5 Yokotani, Seta Oe-cho, Otsu, Shiga 520-2194, Japan
| | - Hiroko Segawa
- Department of Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
24
|
Hu X, Li X, Ye N, Zhou Z, Li G, Jiang F. Association of serum soluble α‑klotho with risk of kidney stone disease: a population-based cross-sectional study. World J Urol 2024; 42:219. [PMID: 38587631 DOI: 10.1007/s00345-024-04837-1] [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: 10/16/2023] [Accepted: 01/16/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND The aim of the study was to explore the association of serum soluble klotho with kidney stone disease (KSD) in the general population over the age of 40 years in the United States. METHODS We integrated the data in National Health and Nutrition Examination Survey from 2007 to 2016 years. The relationship between serum soluble α‑klotho and prevalence of KSD was analyzed by constructing weighted multivariable logistic regression model, restricted cubic spline (RCS) curve, and subgroup analyses. RESULTS In the study, a total of 13,722 individuals were included in our study. A U-shaped association between serum soluble klotho and the risk of KSD was shown by the RCS curve (P value for nonlinear < 0.05). In the full adjusted model, compared with the lowest quartile of serum soluble α‑klotho, the adjusted odd ratios (95% confidence intervals) for KSD across the quartiles were (0.999 (0.859, 1.164), 1.005 (0.858, 1.176), and 1.061 (0.911, 1.235)). Subgroup analyses also showed that the U-shaped association of serum soluble α‑klotho with KSD was found among subjects who were age < 60 years, female or male, with or without hypertension, and BMI ≥ 30 kg/m2. CONCLUSIONS Our findings suggested that serum klotho levels had a U-shaped correlation with risk of KSD. When the Klotho level is at 818.66 pg/mL, prevalence of KSD is lowest. Therefore, maintaining a certain level of serum soluble α‑klotho could prevent the occurrence of KSD.
Collapse
Affiliation(s)
- Xudong Hu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, 100 Huaihai Road, Hefei, 230000, Anhui, China
- Department of Urology, Anhui Public Health Clinical Center, 100 Huaihai Road, Hefei, 230000, Anhui, China
| | - Xiang Li
- Department of Urology, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, 215300, Jiangsu, China
| | - Nan Ye
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, 100 Huaihai Road, Hefei, 230000, Anhui, China
- Department of Urology, Anhui Public Health Clinical Center, 100 Huaihai Road, Hefei, 230000, Anhui, China
| | - Zhenwen Zhou
- Department of Urology, Anqing First People's Hospital of Anhui Province, 42 Xiaosu Road, Anqing, 246000, Anhui, China
| | - Guangyuan Li
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, 100 Huaihai Road, Hefei, 230000, Anhui, China.
- Department of Urology, Anhui Public Health Clinical Center, 100 Huaihai Road, Hefei, 230000, Anhui, China.
| | - Fang Jiang
- Department of Urology, Anqing First People's Hospital of Anhui Province, 42 Xiaosu Road, Anqing, 246000, Anhui, China.
| |
Collapse
|
25
|
Liu J, Wang H, Liu Q, Long S, Wu Y, Wang N, Lin W, Chen G, Lin M, Wen J. Klotho exerts protection in chronic kidney disease associated with regulating inflammatory response and lipid metabolism. Cell Biosci 2024; 14:46. [PMID: 38584258 PMCID: PMC11000353 DOI: 10.1186/s13578-024-01226-4] [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: 10/18/2023] [Accepted: 03/27/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND The anti-aging protein Klotho plays a protective role in kidney disease, but its potential as a biomarker for chronic kidney disease (CKD) is controversial. Additionally, the main pathways through which Klotho exerts its effects on CKD remain unclear. Therefore, we used bioinformatics and clinical data analysis to determine its role in CKD. RESULTS We analyzed the transcriptomic and clinical data from the Nephroseq v5 database and found that the Klotho gene was mainly expressed in the tubulointerstitium, and its expression was significantly positively correlated with estimated glomerular filtration rate (eGFR) and negatively correlated with blood urea nitrogen (BUN) in CKD. We further found that Klotho gene expression was mainly negatively associated with inflammatory response and positively associated with lipid metabolism in CKD tubulointerstitium by analyzing two large sample-size CKD tubulointerstitial transcriptome datasets. By analyzing 10-year clinical data from the National Health and Nutrition Examination Survey (NHANES) 2007-2016, we also found that Klotho negatively correlated with inflammatory biomarkers and triglyceride and positively correlated with eGFR in the CKD population. Mediation analysis showed that Klotho could improve renal function in the general population by modulating the inflammatory response and lipid metabolism, while in the CKD population, it primarily manifested by mediating the inflammatory response. Restricted cubic spline (RCS) analysis showed that the optimal concentration range for Klotho to exert its biological function was around 1000 pg/ml. Kaplan-Meier curves showed that lower cumulative hazards of all-cause mortality in participants with higher levels of Klotho. We also demonstrated that Klotho could reduce cellular inflammatory response and improve cellular lipid metabolism by establishing an in vitro model similar to CKD. CONCLUSIONS Our results suggest that Klotho exerts protection in CKD, which may be mainly related to the regulation of inflammatory response and lipid metabolism, and it can serve as a potential biomarker for CKD.
Collapse
Affiliation(s)
- Junhui Liu
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
- Department of Endocrinology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Huaicheng Wang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
| | - Qinyu Liu
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
- Department of Endocrinology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Shushu Long
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
- Department of Endocrinology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yanfang Wu
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
- Department of Endocrinology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Nengying Wang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
- Department of Endocrinology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Wei Lin
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
- Department of Endocrinology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Gang Chen
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China.
- Department of Endocrinology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China.
| | - Miao Lin
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China.
- Department of Nephrology, Provincial Clinical College, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, China.
| | - Junping Wen
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China.
- Department of Endocrinology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China.
| |
Collapse
|
26
|
Han X, Akinseye L, Sun Z. KDM6A Demethylase Regulates Renal Sodium Excretion and Blood Pressure. Hypertension 2024; 81:541-551. [PMID: 38164755 PMCID: PMC10922853 DOI: 10.1161/hypertensionaha.123.22026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND KDM6A (Lysine-Specific Demethylase 6A) is a specific demethylase for histone 3 lysine (K) 27 trimethylation (H3K27me3). The purpose of this study is to investigate whether KDM6A in renal tubule cells plays a role in the regulation of kidney function and blood pressure. METHODS We first crossed Ksp-Cre+/- and KDM6Aflox/flox mice for generating inducible kidney-specific deletion of KDM6A gene. RESULTS Notably, conditional knockout of KDM6A gene in renal tubule cells (KDM6A-cKO) increased H3K27me3 levels which leads to a decrease in Na excretion and elevation of blood pressure. Further analysis showed that the expression of NKCC2 (Na-K-2Cl cotransporter 2) and NCC (Na-Cl cotransporters) was upregulated which contributes to impaired Na excretion in KDM6A-cKO mice. The expression of AQP2 (aquaporin 2) was also increased in KDM6A-cKO mice, which may facilitate water reabsorption in KDM6A-cKO mice. The expression of Klotho was downregulated while expression of aging markers including p53, p21, and p16 was upregulated in kidneys of KDM6A-cKO mice, indicating that deletion of KDM6A in the renal tubule cells promotes kidney aging. Interestingly, KDM6A-cKO mice developed salt-sensitive hypertension which can be rescued by treatment with Klotho. KDM6A deficiency induced salt-sensitive hypertension likely through downregulation of the Klotho/ERK (extracellular signal-regulated kinase) signaling and upregulation of the WNK (with-no-lysine kinase) signaling. CONCLUSIONS This study provides the first evidence that KDM6A plays an essential role in maintaining normal tubular function and blood pressure. Renal tubule cell specific KDM6A deficiency causes hypertension due to increased H3K27me3 levels and the resultant downregulation of Klotho gene expression which disrupts the Klotho/ERK/NCC/NKCC2 signaling.
Collapse
Affiliation(s)
- Xiaobin Han
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Leah Akinseye
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Zhongjie Sun
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
27
|
Sowaity ZA, Saleem JY, Sabooh TN, Dukmak ON, Abu Al-Saoud SY. Three Siblings With a Rare Familial Hyperphosphatemia Syndrome: A Case Series. Cureus 2024; 16:e55575. [PMID: 38576700 PMCID: PMC10994165 DOI: 10.7759/cureus.55575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
Hyperphosphatemia familial tumoral calcinosis (HFTC) and hyperphosphatemia hyperostosis syndrome (HHS) are rare autosomal recessive disorders caused by mutations in the polypeptide N-acetylgalactosaminyltransferase 3 (GALNT3), fibroblast growth factor 23 (FGF23), or klotho (KL) genes. They are characterized by hyperphosphatemia and recurrent episodes of bone lesions with hyperostosis and/or soft tissue calcinosis. Management options include phosphate-lowering therapies, anti-inflammatory medications, and surgical excision of the calcified masses in significantly disabled cases. We describe three cases from a consanguineous family who were found to have the same genetic mutation caused by a homozygous mutation in intron eight of GALNT3 c.1524+1 G>A (IVS8+1). The first case had a presentation similar to chronic osteomyelitis, while the second one presented with a calcified mass in her gluteal area. The third case presented with left leg pain. Being a rare disease, the findings of tumoral calcinosis/ bony abnormalities, along with elevated phosphate levels, should raise the possibility of this entity. Family history and biochemical findings can help reach the diagnosis.
Collapse
Affiliation(s)
| | | | | | | | - Sima Y Abu Al-Saoud
- Department of Pediatrics, Makassed Hospital, Al-Quds University, Jerusalem, PSE
| |
Collapse
|
28
|
Du T, Liu J, Dong J, Xie H, Wang X, Yang X, Yang Y. Multifunctional coatings of nickel-titanium implant toward promote osseointegration after operation of bone tumor and clinical application: a review. Front Bioeng Biotechnol 2024; 12:1325707. [PMID: 38444648 PMCID: PMC10912669 DOI: 10.3389/fbioe.2024.1325707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 01/29/2024] [Indexed: 03/07/2024] Open
Abstract
Metal implants, especially Ni-Ti shape memory alloy (Ni-Ti SMA) implants, have increasingly become the first choice for fracture and massive bone defects after orthopedic bone tumor surgery. In this paper, the internal composition and shape memory properties of Ni-Ti shape memory alloy were studied. In addition, the effects of porous Ni-Ti SMA on osseointegration, and the effects of surface hydrophobicity and hydrophilicity on the osseointegration of Ni-Ti implants were also investigated. In addition, the effect of surface coating modification technology of Ni-Ti shape memory alloy on bone bonding was also studied. Several kinds of Ni-Ti alloy implants commonly used in orthopedic clinic and their advantages and disadvantages were introduced. The surface changes of Ni-Ti alloy implants promote bone fusion, enhance the adhesion of red blood cells and platelets, promote local tissue regeneration and fracture healing. In the field of orthopaedics, the use of Ni-Ti shape memory alloy implants significantly promoted clinical development. Due to the introduction of the coating, the osseointegration and biocompatibility of the implant surface have been enhanced, and the success rate of the implant has been greatly improved.
Collapse
Affiliation(s)
- Tianhao Du
- Department of Rehabilitation Medicine, General Hospital of Northern Theater Command, Shenyang, China
- Liaoning University of traditional Chinese Medicine, Shenyang, China
| | - Jia Liu
- Liaoning University of traditional Chinese Medicine, Shenyang, China
| | - Jinhan Dong
- Liaoning University of traditional Chinese Medicine, Shenyang, China
| | - Haoxu Xie
- Department of Rehabilitation Medicine, General Hospital of Northern Theater Command, Shenyang, China
- Liaoning University of traditional Chinese Medicine, Shenyang, China
| | - Xiao Wang
- Department of Rehabilitation Medicine, General Hospital of Northern Theater Command, Shenyang, China
- Liaoning University of traditional Chinese Medicine, Shenyang, China
| | - Xu Yang
- Liaoning University of traditional Chinese Medicine, Shenyang, China
| | - Yingxin Yang
- Liaoning University of traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
29
|
Dusso A, Bauerle KT, Zhang RM, Bernal-Mizrachi C. Vitamin D and renal disease. FELDMAN AND PIKE'S VITAMIN D 2024:587-618. [DOI: 10.1016/b978-0-323-91338-6.00029-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
30
|
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: 47] [Impact Index Per Article: 47.0] [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
|
31
|
Rostamzadeh F, Moosavi-Saeed Y, Yeganeh-Hajahmadi M. Interaction of Klotho and sirtuins. Exp Gerontol 2023; 182:112306. [PMID: 37804921 DOI: 10.1016/j.exger.2023.112306] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 09/30/2023] [Accepted: 10/02/2023] [Indexed: 10/09/2023]
Abstract
OBJECTIVE In this article, we review the articles that have reported the interaction between Klotho and sirtuins. RECENT FINDINGS Sirtuins are a family of histone deacetylase enzymes that are considered to be the main regulators of biological processes. This family is one of the essential factors for postponing aging and increasing the life span of organisms. Sirtuins play a role in regulating the function of various cellular processes such as cellular metabolism, oxidative stress, apoptosis, and inflammation. It has also been shown that various diseases are related to these enzymes. Klotho is an anti-aging protein that exists as a membrane protein as well as a soluble circulating form. The membrane type of this protein acts as a co-receptor of the FGF endocrine family. It has been shown that the Klotho gene is related to age-related diseases, including osteoporosis, coronary artery, brain diseases, diabetes, etc. At the same time, it is difficult to separate the actions of Klotho and endocrine FGFs. Several studies have shown that Klotho and sirtuins interact with each other at different regulatory levels. However, it is necessary to carry out more in-vivo investigations to create new windows towards the treatment or prevention of various diseases.
Collapse
Affiliation(s)
- Farzaneh Rostamzadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Yasamin Moosavi-Saeed
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahboobeh Yeganeh-Hajahmadi
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Science, Kerman, Iran.
| |
Collapse
|
32
|
Zhang Y, Zhao C, Zhang H, Chen M, Meng Y, Pan Y, Zhuang Q, Zhao M. Association between serum soluble α-klotho and bone mineral density (BMD) in middle-aged and older adults in the United States: a population-based cross-sectional study. Aging Clin Exp Res 2023; 35:2039-2049. [PMID: 37368163 DOI: 10.1007/s40520-023-02483-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/18/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND Osteoporosis is a degenerative disease defined by low bone mineral density, has a high prevalence, and causes fractures at multiple sites throughout the body, greatly affecting the quality of patients. α-Klotho is an endocrine factor involved in the regulation of various metabolic processes in humans, and its role in bone metabolism has attracted widespread attention. The relationship between α-klotho and bone mineral density has not been uniformly recognized, and no large-scale correlation analysis has been conducted in the middle-aged and elderly population. OBJECTIVE To determine the relationship between α-klotho and bone mineral density in middle-aged and elderly people. METHODS Population data of 3120 individuals aged 40-79 years were obtained from the NHANES database for the period 2011-2016. Regression analysis was performed using a general linear model with serum α-klotho as the independent variable and total bone mineral density, thoracic bone mineral density, lumbar bone mineral density, pelvic bone mineral density, and trunk bone mineral density as the dependent variables, respectively. The generalized additive model was also used for smoothing curve fitting and threshold effect analysis. RESULTS Serum α-klotho was positively correlated with total bone mineral density at lg (Klotho) < 2.97 and with thoracic bone mineral density at lg (Klotho) > 2.69 (β = 0.05, p = 0.0006), and negatively correlated (β = -0.27, p = 0.0341) with lumbar bone mineral density at lg (Klotho) < 2.69. It also positively correlated with trunk bone mineral density (β = 0.027, p = 0.03657) and had no segmental effect but did not correlate with pelvic bone mineral density. The positive association of serum α-klotho with those aged 40-49 years, female, non-Hispanic White, and without hypertension was clearer. In the population with diabetes, a significantly positive association between total (β = 0.15, p = 0.01), thoracic (β = 0.23, p = 0.0404), and lumbar (β = 0.22, p = 0.0424) bone mineral density and α-klotho was observed. CONCLUSIONS α-Klotho has different relationships with total, thoracic, lumbar, and trunk bone mineral density. Among them, the positive correlation between α-klotho and trunk bone mineral density is more valuable for predicting osteoporosis. The significant effect of α-klotho on bone mineral density in diabetes patients suggests its potential as a predictive marker of diabetes progression.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Changtai Zhao
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Hanyong Zhang
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, 410219, China
| | - Mingcong Chen
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Yang Meng
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Yuxin Pan
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Quan Zhuang
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
33
|
Jiang J, Liu Q, Mao Y, Wang N, Lin W, Li L, Liang J, Chen G, Huang H, Wen J. Klotho reduces the risk of osteoporosis in postmenopausal women: a cross-sectional study of the National Health and Nutrition Examination Survey (NHANES). BMC Endocr Disord 2023; 23:151. [PMID: 37452417 PMCID: PMC10347835 DOI: 10.1186/s12902-023-01380-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 05/22/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Osteoporosis (OP) is one of the diseases that endanger the health of the elderly population. Klotho protein is a hormone with anti-aging effects. A few studies have discussed the relationship between Klotho and OP. However, there is still a lack of research on larger populations. This study aims to evaluate the association between OP and Klotho in American postmenopausal women. METHODS This is a retrospective study. We searched the National Health and Nutrition Examination Survey (NHANES) database and collected data of 3 survey cycles, finally involving 871 postmenopausal women over 50 years old in the present study. All participants took dual-energy X-ray absorptiometry examination and serum Klotho testing at the time of investigation. After adjusting the possible confounding variables, a multivariate regression model was employed to estimate the relationship between OP and Klotho proteins. Besides, the P for trend and restricted cubic spline (RCS) were applied to examine the threshold effect and calculate the inflection point. RESULTS Factors influencing the occurrence of OP included age, ethnicity, body mass index and Klotho levels. Multivariate regression analysis indicated that the serum Klotho concentration was lower in OP patients than that in participants without OP (OR[log2Klotho] = 0.568, P = 0.027). The C-index of the prediction model built was 0.765, indicating good prediction performance. After adjusting the above-mentioned four variables, P values for trend showed significant differences between groups. RCSs revealed that when the Klotho concentration reached 824.09 pg/ml, the risk of OP decreased drastically. CONCLUSION Based on the analysis of the data collected from the NHANES database, we propose a correlation between Klotho and postmenopausal OP. A higher serum Klotho level is related to a lower incidence of OP. The findings of the present study can provide guidance for research on diagnosis and risk assessment of OP.
Collapse
Affiliation(s)
- Jialin Jiang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Endocrinology, Fujian Provincial Hospital, Fuzhou, China
| | - Qinyu Liu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Endocrinology, Fujian Provincial Hospital, Fuzhou, China
| | - Yaqian Mao
- Department of Internal Medicine, Fujian Provincial Hospital Jinshan Branch, Fuzhou, China
| | - Nengyin Wang
- Department of Endocrinology, Fujian Provincial Hospital, Fuzhou, China
| | - Wei Lin
- Department of Endocrinology, Fujian Provincial Hospital, Fuzhou, China
| | - Liantao Li
- Department of Endocrinology, Fujian Provincial Hospital, Fuzhou, China
| | - Jixing Liang
- Department of Endocrinology, Fujian Provincial Hospital, Fuzhou, China
| | - Gang Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Endocrinology, Fujian Provincial Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Medical Analysis, Fujian Academy of Medical, Fuzhou, China
| | - Huibin Huang
- Department of Endocrinology, Fujian Provincial Hospital, Fuzhou, China.
| | - Junping Wen
- Department of Endocrinology, Fujian Provincial Hospital, Fuzhou, China.
| |
Collapse
|
34
|
Liu S, Wu M, Wang Y, Xiang L, Luo G, Lin Q, Xiao L. The Association between Dietary Fiber Intake and Serum Klotho Levels in Americans: A Cross-Sectional Study from the National Health and Nutrition Examination Survey. Nutrients 2023; 15:3147. [PMID: 37513564 PMCID: PMC10385840 DOI: 10.3390/nu15143147] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/28/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Klotho is an aging-related marker closely associated with a number of diseases. A growing body of evidence suggests that dietary factors and lifestyle habits can impact serum Klotho levels. The effect of dietary fiber, a key component of a healthy diet, on the body's serum Klotho levels has not been fully elucidated. OBJECTIVE The aim of this study was to explore the relationship between dietary fiber intake and serum Klotho levels in people aged 40-79 years in the United States. METHODS A total of 11,282 participants were included in this study, all from the National Health and Nutrition Examination Survey from 2007 to 2016. Dietary fiber intake was assessed by uniformly trained interviewers using the 24 h dietary recall method. Serum Klotho was quantified using commercially available ELISA kits manufactured by IBL International, Japan. The relationship between dietary fiber intake and serum Klotho levels was analyzed using a multiple linear regression model. Subsequently, the non-linear dose-response relationship between the two was further explored using a restricted cubic spline (RCS) model. RESULTS After adjusting for potential confounders, serum Klotho levels increased by 1.9% (95% confidence interval [CI]: 0.8%, 3.0%) for each interquartile range increase in dietary fiber intake in all participants. Considering dietary fiber intake as a categorical variable, serum Klotho levels were found to be 4.7% higher in participants in the highest quartile of dietary fiber intake than in those in the lowest quartile (95% CI: 1.8%, 7.6%). RCS plots depicted a non-linear positive correlation between dietary fiber intake and serum Klotho levels. Subgroup analysis revealed that the relationship between dietary fiber intake and serum Klotho levels was more pronounced in older (percentage change: 7.0%; 95% CI: 2.5%, 11.7%) and overweight and obese participants (percentage change: 4.9%; 95% CI: 1.5%, 8.4%). CONCLUSIONS The results of this study showed that dietary fiber intake was significantly associated with serum Klotho levels in participants. This finding is yet to be further confirmed by prospective studies.
Collapse
Affiliation(s)
- Si Liu
- Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Mingyang Wu
- Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Yan Wang
- Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Lu Xiang
- Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Gang Luo
- Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Qian Lin
- Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Lin Xiao
- Xiangya School of Public Health, Central South University, Changsha 410078, China
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Fibroblast growth factor (FGF) 23 is a bone-derived hormone that regulates phosphate and vitamin D metabolism by targeting the kidney. When highly elevated, such as in chronic kidney disease (CKD), FGF23 can also target the heart and induce pathologic remodeling. Here we discuss the mechanisms that underlie the physiologic and pathologic actions of FGF23, with focus on its FGF receptors (FGFR) and co-receptors. RECENT FINDINGS Klotho is a transmembrane protein that acts as an FGFR co-receptor for FGF23 on physiologic target cells. Klotho also exists as a circulating variant, and recent studies suggested that soluble klotho (sKL) can mediate FGF23 effects in cells that do not express klotho. Furthermore, it has been assumed that the actions of FGF23 do not require heparan sulfate (HS), a proteoglycan that acts as a co-receptor for other FGF isoforms. However, recent studies revealed that HS can be part of the FGF23:FGFR signaling complex and modulate FGF23-induced effects. SUMMARY sKL and HS have appeared as circulating FGFR co-receptors that modulate the actions of FGF23. Experimental studies suggest that sKL protects from and HS accelerates CKD-associated heart injury. However, the in vivo relevance of these findings is still speculative.
Collapse
Affiliation(s)
- S Madison Thomas
- Division of Nephrology and Section of Mineral Metabolism, Department of Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | |
Collapse
|
36
|
Cai Q, Hu S, Qi C, Yin J, Xu S, Hou FF, Li A. Serum Anti-Aging Protein α-Klotho Mediates the Association between Diet Quality and Kidney Function. Nutrients 2023; 15:2744. [PMID: 37375648 PMCID: PMC10301566 DOI: 10.3390/nu15122744] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/08/2023] [Accepted: 06/11/2023] [Indexed: 06/29/2023] Open
Abstract
Adherence to healthy dietary patterns is associated with a reduced risk of kidney dysfunction. Nevertheless, the age-related mechanisms that underpin the relationship between diet and kidney function remain undetermined. This study aimed to investigate the mediating role of serum α-Klotho, an anti-aging protein, in the link between a healthy diet and kidney function. A cross-sectional study was conducted on a cohort of 12,817 individuals aged between 40 and 79 years who participated in the National Health and Nutrition Examination Survey (NHANES) from 2007 to 2016. For each participant, the Healthy Eating Index 2015 (HEI-2015) score was calculated as a measure of a healthy dietary pattern. Creatinine-based estimated glomerular filtration rate (eGFR) was used to assess kidney function. Multivariable regression models were used to analyze the association between the standardized HEI-2015 score and eGFR after adjusting for potential confounders. Causal mediation analysis was performed to assess whether serum α-Klotho influenced this association. The mean (±SD) eGFR of all individuals was 86.8 ± 19.8 mL/min per 1.73 m2. A high standardized HEI-2015 score was associated with a high eGFR (β [95% CI], 0.94 [0.64-1.23]; p < 0.001). The mediation analysis revealed that serum α-Klotho accounted for 5.6-10.5% of the association of standardized overall HEI-2015 score, total fruits, whole fruits, greens and beans, and whole grain with eGFR in the NHANES. According to the results from the subgroup analysis, serum α-Klotho exerted a mediating effect in the participants aged 60-79 years and in males. A healthy diet may promote kidney function by up-regulating serum anti-aging α-Klotho. This novel pathway suggests important implications for dietary recommendations and kidney health.
Collapse
Affiliation(s)
- Qingqing Cai
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China;
| | - Shixian Hu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China;
- Department of Gastroenterology and Hepatology, University Medical Center Groningen (UMCG), University of Groningen, 9712 Groningen, The Netherlands
| | - Cancan Qi
- Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China;
| | - Jiawei Yin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety & Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China;
| | - Shulan Xu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China;
| | - Fan Fan Hou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou 510515, China;
| | - An Li
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China;
| |
Collapse
|
37
|
Aihemaitijiang B, Ruotian L, Qi Y, Mahemuti M. Correlation between sKL and Nrf2 plasma levels and calcium oxalate urolithiasis. Int Urol Nephrol 2023:10.1007/s11255-023-03615-z. [PMID: 37198517 DOI: 10.1007/s11255-023-03615-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/23/2023] [Indexed: 05/19/2023]
Abstract
OBJECTIVE To investigate the relationship between plasma levels of sKL and Nrf2 and calcium oxalate calculi. METHODS The clinical data of 135 patients with calcium oxalate calculi treated in the Department of Urology of the second affiliated Hospital of Xinjiang Medical University from February 2019 to December 2022, and 125 healthy persons who underwent physical examination in the same period were collected and divided into healthy group and stone group. The levels of sKL and Nrf2 were measured by ELISA. Correlation test was used to analyze the risk factors of calcium oxalate stones, logistic regression analysis was used to analyze the risk factors of calcium oxalate stones, and ROC curve was used to evaluate the sensitivity and specificity of sKL and Nrf2 in predicting urinary calculi. RESULTS Compared with the healthy group, the plasma sKL level in the stone group decreased (111.53 ± 27.89 vs 130.68 ± 32.51), while the plasma Nrf2 level increased (300.74 ± 114.31 vs 246.74 ± 108.22). There was no significant difference in the distribution of age and sex between the healthy group and the stone group, but there were significant differences in plasma levels of WBC, NEUT, CRP, BUN, BUA, SCr, BMI, and eating habits. The results of correlation test showed that the level of plasma Nrf2 was positively correlated with SCr (r = 0.181, P < 0.05) and NEUT (r = 0.144 P < 0.05). Plasma sKL was not significantly correlated with Nrf2 (r = 0.047, P > 0.05), WBC (r = 0.108, P > 0.05), CRP (r = - 0.022, P > 0.05), BUN (r = - 0.115, P > 0.05), BUA (r = - 0.139, P > 0.05), SCr (r = 0.049, P > 0.05), and NEUT (r = 0.027, P > 0.05). Plasma Nrf2 was not significantly correlated with WBC (r = 0.097, P > 0.05), CRP (r = 0.045, P > 0.05), BUN (r = 0.122, P > 0.05), and BUA (r = 0.122, P > 0.05); (r = 0.078, P > 0.05) had no significant correlation. Logistic regression showed that elevated plasma sKL (OR 0.978, 95% CI 0.969 ~ 0.988, P < 0.05) was a protective factor for the occurrence of calcium oxalate stones, BMI (OR 1.122, 95% CI 1.045 ~ 1.206, P < 0.05), dietary habit score (OR 1.571, 95% CI 1.221 ~ 2.020, P < 0.05), and WBC (OR 1.551, 95% CI 1.423 ~ 1.424, P < 0.05). Increased NEUT (OR 1.539, 95% CI 1.391 ~ 1.395, P < 0.05) and CRP (OR 1.118, 95% CI: 1.066 ~ 1.098, P < 0.05) are risk factors for the occurrence of calcium oxalate stones. CONCLUSION Plasma sKL level decreased and Nrf2 level increased in patients with calcium oxalate calculi. Plasma sKL may play an antioxidant role in the pathogenesis of calcium oxalate stones through Nrf2 antioxidant pathway.
Collapse
Affiliation(s)
- Baihetiyaer Aihemaitijiang
- Urology Department, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Liu Ruotian
- Department of Urology, The Second People's Hospital of Henan Province, Zhengzhou, 450000, Henan Province, China
| | - Yang Qi
- Department of Surgery, Neijiang Traditional Chinese Medicine Hospital, Neijiang, 641099, Sichuan, China
| | - Mulati Mahemuti
- Urology Department, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China.
- Department of Urology, The Second People's Hospital of Henan Province, Zhengzhou, 450000, Henan Province, China.
- Department of Surgery, Neijiang Traditional Chinese Medicine Hospital, Neijiang, 641099, Sichuan, China.
| |
Collapse
|
38
|
Li M, Ma Y, Cheng W, Zhang L, Zhou C, Zhang W, Zhang W. Association between perfluoroalkyl and polyfluoroalkyl internal exposure and serum α-Klotho levels in middle-old aged participants. Front Public Health 2023; 11:1136454. [PMID: 37228732 PMCID: PMC10204767 DOI: 10.3389/fpubh.2023.1136454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 04/11/2023] [Indexed: 05/27/2023] Open
Abstract
Purpose Exposure to perfluoroalkyl and polyfluoroalkyl substances causes oxidative stress, which is strongly associated with adverse health effects. Klotho protein plays an anti-aging role via antioxidation activity. Methods We investigated the levels of serum α-Klotho and PFAS exposure in adults who participated in the National Health and Nutrition Examination Survey from 2013 to 2016. A nationally representative subsample of 1,499 adults aged 40-79 years was analyzed for the associations of serum α-Klotho levels with serum PFAS exposures by correlation analysis and multiple general linear models. Of note, the potential confounding factors including age and gender were adjusted. Quantile-based g-computation models were used to assess the effects of mixed PFAS exposure on serum α-Klotho levels. Results The weighted geometric mean of serum α-Klotho was 791.38 pg/mL for the subjects during 2013-2016. After adjusting for potential confounders, serum Klotho levels showed a statistically significant downward trend with increasing quartiles of PFOA and PFNA. Multivariate adjusted general linear regression analysis showed that increased exposure to PFNA was substantially associated with lower serum levels of α-Klotho, and each 1-unit increase in PFNA concentration was accompanied by a 20.23 pg/mL decrease in α-Klotho level; while no significant association was observed between other PFAS exposures and serum α-Klotho levels. It was negatively correlated between α-Klotho and Q4 for PFNA relative to the lowest quartile (Q1) of exposure (P = 0.025). It was found that the strongest negative correlation between PFNA exposure and serum α-Klotho levels was in the middle-aged (40-59 years) female participants. Furthermore, the mixture of the four PFAS substances showed an overall inverse association with serum α-Klotho concentrations, with PFNA being the major contributor. Conclusions Taken together, in a representative sample of the U.S. middle-aged and elderly populations, serum concentrations of PFAS, especially PFNA, have been negatively associated with serum levels of α-Klotho, which is strongly associated with cognition and aging. It was important to note that the majority of associations were limited to middle-aged women. It will be meaningful to clarify the causal relationship and the pathogenic mechanisms of PFAS exposure and α-Klotho levels, which is helpful to aging and aging-related diseases.
Collapse
Affiliation(s)
- Min Li
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
- Guangdong Provincial Engineering and Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Yuanlin Ma
- Reproductive Medicine Center, Guangdong Provincial Key Laboratory of Reproductive Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenli Cheng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Luyun Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Cheng Zhou
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Wenji Zhang
- Guangdong Provincial Engineering and Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| | - Wenjuan Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
39
|
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
|
40
|
Martín-Carro B, Martín-Vírgala J, Fernández-Villabrille S, Fernández-Fernández A, Pérez-Basterrechea M, Navarro-González JF, Donate-Correa J, Mora-Fernández C, Dusso AS, Carrillo-López N, Panizo S, Naves-Díaz M, Fernández-Martín JL, Cannata-Andía JB, Alonso-Montes C. Role of Klotho and AGE/RAGE-Wnt/β-Catenin Signalling Pathway on the Development of Cardiac and Renal Fibrosis in Diabetes. Int J Mol Sci 2023; 24:5241. [PMID: 36982322 PMCID: PMC10049403 DOI: 10.3390/ijms24065241] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Fibrosis plays an important role in the pathogenesis of long-term diabetic complications and contributes to the development of cardiac and renal dysfunction. The aim of this experimental study, performed in a long-term rat model, which resembles type 1 diabetes mellitus, was to investigate the role of soluble Klotho (sKlotho), advanced glycation end products (AGEs)/receptor for AGEs (RAGE), fibrotic Wnt/β-catenin pathway, and pro-fibrotic pathways in kidney and heart. Diabetes was induced by streptozotocin. Glycaemia was maintained by insulin administration for 24 weeks. Serum and urine sKlotho, AGEs, soluble RAGE (sRAGE) and biochemical markers were studied. The levels of Klotho, RAGEs, ADAM10, markers of fibrosis (collagen deposition, fibronectin, TGF-β1, and Wnt/β-catenin pathway), hypertrophy of the kidney and/or heart were analysed. At the end of study, diabetic rats showed higher levels of urinary sKlotho, AGEs and sRAGE and lower serum sKlotho compared with controls without differences in the renal Klotho expression. A significant positive correlation was found between urinary sKlotho and AGEs and urinary albumin/creatinine ratio (uACR). Fibrosis and RAGE levels were significantly higher in the heart without differences in the kidney of diabetic rats compared to controls. The results also suggest the increase in sKlotho and sRAGE excretion may be due to polyuria in the diabetic rats.
Collapse
Affiliation(s)
- Beatriz Martín-Carro
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Julia Martín-Vírgala
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sara Fernández-Villabrille
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | - Marcos Pérez-Basterrechea
- Unit of Cell Therapy and Regenerative Medicine, Hematology and Hemotherapy Service, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Juan F. Navarro-González
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
- Nephrology Service, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
| | - Javier Donate-Correa
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
| | - Carmen Mora-Fernández
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
| | - Adriana S. Dusso
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Natalia Carrillo-López
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sara Panizo
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Manuel Naves-Díaz
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José L. Fernández-Martín
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jorge B. Cannata-Andía
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Medicine, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Cristina Alonso-Montes
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Psychobiology, Universidad de Oviedo, 33003 Oviedo, Spain
| |
Collapse
|
41
|
Regulation of FGF23 production and phosphate metabolism by bone-kidney interactions. Nat Rev Nephrol 2023; 19:185-193. [PMID: 36624273 DOI: 10.1038/s41581-022-00665-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2022] [Indexed: 01/11/2023]
Abstract
The bone-derived hormone fibroblast growth factor 23 (FGF23) functions in concert with parathyroid hormone (PTH) and the active vitamin D metabolite, 1,25(OH)2 vitamin D (1,25D), to control phosphate and calcium homeostasis. A rise in circulating levels of phosphate and 1,25D leads to FGF23 production in bone. Circulating FGF23 acts on the kidney by binding to FGF receptors and the co-receptor α-Klotho to promote phosphaturia and reduce circulating 1,25D levels. Various other biomolecules that are produced by the kidney, including lipocalin-2, glycerol 3-phosphate, 1-acyl lysophosphatidic acid and erythropoietin, are involved in the regulation of mineral metabolism via effects on FGF23 synthesis in bone. Understanding of the molecular mechanisms that control FGF23 synthesis in the bone and its bioactivity in the kidney has led to the identification of potential targets for novel interventions. Emerging approaches to target aberrant phosphate metabolism include small molecule inhibitors that directly bind FGF23 and prevent its interactions with FGF receptors and α-Klotho, FGF23 peptide fragments that act as competitive inhibitors of intact FGF23 and small molecule inhibitors of kidney sodium-phosphate cotransporters.
Collapse
|
42
|
Soluble Klotho protects against glomerular injury through regulation of ER stress response. Commun Biol 2023; 6:208. [PMID: 36813870 PMCID: PMC9947099 DOI: 10.1038/s42003-023-04563-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
αKlotho (Klotho) has well established renoprotective effects; however, the molecular pathways mediating its glomerular protection remain incompletely understood. Recent studies have reported that Klotho is expressed in podocytes and protects glomeruli through auto- and paracrine effects. Here, we examined renal expression of Klotho in detail and explored its protective effects in podocyte-specific Klotho knockout mice, and by overexpressing human Klotho in podocytes and hepatocytes. We demonstrate that Klotho is not significantly expressed in podocytes, and transgenic mice with either a targeted deletion or overexpression of Klotho in podocytes lack a glomerular phenotype and have no altered susceptibility to glomerular injury. In contrast, mice with hepatocyte-specific overexpression of Klotho have high circulating levels of soluble Klotho, and when challenged with nephrotoxic serum have less albuminuria and less severe kidney injury compared to wildtype mice. RNA-seq analysis suggests an adaptive response to increased endoplasmic reticulum stress as a putative mechanism of action. To evaluate the clinical relevance of our findings, the results were validated in patients with diabetic nephropathy, and in precision cut kidney slices from human nephrectomies. Together, our data reveal that the glomeruloprotective effects of Klotho is mediated via endocrine actions, which increases its therapeutic potential for patients with glomerular diseases.
Collapse
|
43
|
Ren JR, Wang Z, Cheng Y, He CY, Jian JM, Fan DY, Shen YY, Chen DW, Li HY, Yi X, Zeng GH, Tan CR, Shi AY, Chen LY, Mao QX, Wang YJ, Wang J. Associations Between Plasma Klotho with Renal Function and Cerebrospinal Fluid Amyloid-β Levels in Alzheimer's Disease: The Chongqing Ageing & Dementia Study. J Alzheimers Dis 2023; 92:477-485. [PMID: 36776069 DOI: 10.3233/jad-221107] [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: 02/12/2023]
Abstract
BACKGROUND The kidney-brain crosstalk has been involved in Alzheimer's disease (AD) with the mechanism remaining unclear. The anti-aging factor Klotho was reported to attenuate both kidney injury and AD pathologies. OBJECTIVE To investigate whether plasma Klotho participated in kidney-brain crosstalk in AD. METHODS We enrolled 33 PiB-PET-positive AD patients and 33 amyloid-β (Aβ)-negative age- and sex-matched cognitively normal (CN) controls from the Chongqing Ageing & Dementia Study (CADS). The levels of plasma Klotho, Aβ, and tau in the cerebrospinal fluid (CSF) were measured by enzyme-linked immunosorbent assay. RESULTS We found higher plasma Klotho and lower estimated glomerular filtration rate (eGFR) levels in AD patients compared with CN. The eGFR were positively associated with Aβ 42, Aβ 40 levels in CSF and negatively associated with CSF T-tau levels. Plasma Klotho levels were both negatively correlated with CSF Aβ 42 and eGFR. Mediation analysis showed that plasma Klotho mediated 24.96% of the association between eGFR and CSF Aβ 42. CONCLUSION Renal function impacts brain Aβ metabolism via the kidney-brain crosstalk, in which the plasma klotho may be involved as a mediator. Targeting Klotho to regulate the kidney-brain crosstalk provides potential therapeutic approaches for AD.
Collapse
Affiliation(s)
- Jun-Rong Ren
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Zhen Wang
- Department of Critical Care Medicine, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yuan Cheng
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Chen-Yang He
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Jie-Ming Jian
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Dong-Yu Fan
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Ying-Ying Shen
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Dong-Wan Chen
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Hui-Yun Li
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Xu Yi
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Gui-Hua Zeng
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Cheng-Rong Tan
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - An-Yu Shi
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Li-Yong Chen
- Department of Anaesthesiology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Qing-Xiang Mao
- Department of Anaesthesiology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.,Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| | - Jun Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Institute of Brain and Intelligence, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| |
Collapse
|
44
|
Williams MJ, White SC, Joseph Z, Hruska KA. Updates in the chronic kidney disease-mineral bone disorder show the role of osteocytic proteins, a potential mechanism of the bone-Vascular paradox, a therapeutic target, and a biomarker. Front Physiol 2023; 14:1120308. [PMID: 36776982 PMCID: PMC9909112 DOI: 10.3389/fphys.2023.1120308] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/17/2023] [Indexed: 01/27/2023] Open
Abstract
The chronic kidney disease-mineral bone disorder (CKD-MBD) is a complex multi-component syndrome occurring during kidney disease and its progression. Here, we update progress in the components of the syndrome, and synthesize recent investigations, which suggest a potential mechanism of the bone-vascular paradox. The discovery that calcified arteries in chronic kidney disease inhibit bone remodeling lead to the identification of factors produced by the vasculature that inhibit the skeleton, thus providing a potential explanation for the bone-vascular paradox. Among the factors produced by calcifying arteries, sclerostin secretion is especially enlightening. Sclerostin is a potent inhibitor of bone remodeling and an osteocyte specific protein. Its production by the vasculature in chronic kidney disease identifies the key role of vascular cell osteoblastic/osteocytic transdifferentiation in vascular calcification and renal osteodystrophy. Subsequent studies showing that inhibition of sclerostin activity by a monoclonal antibody improved bone remodeling as expected, but stimulated vascular calcification, demonstrate that vascular sclerostin functions to brake the Wnt stimulation of the calcification milieu. Thus, the target of therapy in the chronic kidney disease-mineral bone disorder is not inhibition of sclerostin function, which would intensify vascular calcification. Rather, decreasing sclerostin production by decreasing the vascular osteoblastic/osteocytic transdifferentiation is the goal. This might decrease vascular calcification, decrease vascular stiffness, decrease cardiac hypertrophy, decrease sclerostin production, reduce serum sclerostin and improve skeletal remodeling. Thus, the therapeutic target of the chronic kidney disease-mineral bone disorder may be vascular osteoblastic transdifferentiation, and sclerostin levels may be a useful biomarker for the diagnosis of the chronic kidney disease-mineral bone disorder and the progress of its therapy.
Collapse
Affiliation(s)
- Matthew J. Williams
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University, Saint Louis, MO, United States
| | - Sarah C. White
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University, Saint Louis, MO, United States
| | - Zachary Joseph
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University, Saint Louis, MO, United States
| | - Keith A. Hruska
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University, Saint Louis, MO, United States
- Departments of Medicine and Cell Biology, Washington University, Saint Louis, MO, United States
| |
Collapse
|
45
|
Barbuto S, Perrone V, Veronesi C, Dovizio M, Zappulo F, Vetrano D, Giannini S, Fusaro M, Ancona DD, Barbieri A, Ferrante F, Lena F, Palcic S, Re D, Rizzi FV, Cogliati P, Soro M, Esposti LD, Cianciolo G. Real-World Analysis of Outcomes and Economic Burden in Patients with Chronic Kidney Disease with and without Secondary Hyperparathyroidism among a Sample of the Italian Population. Nutrients 2023; 15:nu15020336. [PMID: 36678208 PMCID: PMC9867108 DOI: 10.3390/nu15020336] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/06/2023] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
This real-world analysis evaluated the clinical and economic burden of non-dialysis-dependent CKD patients with and without secondary hyperparathyroidism (sHPT) in Italy. An observational retrospective study was conducted using administrative databases containing a pool of healthcare entities covering 2.45 million health-assisted individuals. Adult patients with hospitalization discharge diagnoses for CKD stages 3, 4, and 5 were included from 1 January 2012 to 31 March 2015 and stratified using the presence/absence of sHPT. Of the 5710 patients, 3119 were CKD-only (62%) and 1915 were CKD + sHPT (38%). The groups were balanced using Propensity Score Matching (PSM). Kaplan-Meier curves revealed that progression to dialysis and cumulative mortality had a higher incidence in the CKD + sHPT versus CKD-only group in CKD stage 3 patients and the overall population. The total direct healthcare costs/patient at one-year follow-up were significantly higher in CKD + sHPT versus CKD-only patients (EUR 8593 vs. EUR 5671, p < 0.001), mostly burdened by expenses for drugs (EUR 2250 vs. EUR 1537, p < 0.001), hospitalizations (EUR 4628 vs. EUR 3479, p < 0.001), and outpatient services (EUR 1715 vs. EUR 654, p < 0.001). These findings suggest that sHPT, even at an early CKD stage, results in faster progression to dialysis, increased mortality, and higher healthcare expenditures, thus indicating that timely intervention can ameliorate the management of CKD patients affected by sHPT.
Collapse
Affiliation(s)
- Simona Barbuto
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Valentina Perrone
- CliCon S.r.l., Società Benefit, Health, Economics & Outcomes Research, 40137 Bologna, Italy
| | - Chiara Veronesi
- CliCon S.r.l., Società Benefit, Health, Economics & Outcomes Research, 40137 Bologna, Italy
| | - Melania Dovizio
- CliCon S.r.l., Società Benefit, Health, Economics & Outcomes Research, 40137 Bologna, Italy
| | - Fulvia Zappulo
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Daniele Vetrano
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Sandro Giannini
- Clinica Medica 1, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Maria Fusaro
- National Research Council (CNR), Institute of Clinical Physiology (IFC), 56124 Pisa, Italy
- Department of Medicine, University of Padova, 35128 Padova, Italy
| | | | | | - Fulvio Ferrante
- UOC Farmacia, Ufficio di Farmacovigilanza, ASL Frosinone, 03100 Frosinone, Italy
| | - Fabio Lena
- U.O.C. Politiche del Farmaco, USL Toscana Sud Est, 58100 Grosseto, Italy
| | - Stefano Palcic
- SC Farmacia Ospedaliera e Territoriale—Area Giuliana, Azienda Sanitaria Universitaria Integrata Giuliano-Isontina (ASUGI), 34128 Trieste, Italy
| | - Davide Re
- Servizio Farmaceutico Territoriale, ASL Teramo, 64100 Teramo, Italy
| | | | | | | | - Luca Degli Esposti
- CliCon S.r.l., Società Benefit, Health, Economics & Outcomes Research, 40137 Bologna, Italy
- Correspondence:
| | - Giuseppe Cianciolo
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
46
|
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
|
47
|
Abstract
BACKGROUND & AIMS Soluble α-Klotho (s-Klotho) is a circulating protein with pleiotropic effects that mainly induce protective effects. Our study investigates the associations between s-Klotho and several established inflammatory biomarkers, with the aim of examining whether s-Klotho levels are representative of inflammatory states. METHODS A total of 11,128 eligible participants from NHANES 2007-2016 were included in our study. Levels of four inflammatory biomarkers, uric acid (UA), C-reactive protein (CRP), white blood cell (WBC) count, and mean platelet volume (MPV), were examined for their relationship with s-Klotho levels. Sub-analyses sorted the total population by gender and into four quartiles. Linear regression models were used to evaluate the strengths of associations. RESULTS All four inflammatory biomarkers were significantly associated with s-Klotho levels. UA, CRP, and WBC count showed an inverse association, while MPV showed a direct one. Of the four markers, UA was most strongly correlated with s-Klotho levels (β coefficient: -28.89 in unadjusted model, p<.001), and this relationship was stronger in women than in men (β coefficient of UA in men: -22.01, p<.001; in women: -31.54, p<.001). In addition, all four biomarkers manifested stronger associations with s-Klotho in higher quartiles, and the highest absolute values of β coefficients appeared in Q4 vs. Q1. CONCLUSION s-Klotho is significantly associated with well-recognized inflammatory biomarkers. A decrease in s-Klotho levels implies a general inflammatory status; therefore, s-Klotho serves as a potential biomarker that is inversely correlated with inflammatory conditions. Further applications in clinical practice will provide us with a better understanding of its role.Key messagesSoluble α-Klotho (s-Klotho) levels are significantly associated with the inflammatory markers uric acid, C-reactive protein, white blood cell count, and mean platelet volume.S-Klotho is involved in inflammatory processes and plays a protective role.S-Klotho may serve as an inverse indicator of inflammation.
Collapse
Affiliation(s)
- Shou-En Wu
- Department of Dermatology, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei City, Taiwan
| | - Wei-Liang Chen
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei City, Taiwan.,Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei City, Taiwan.,Department of Biochemistry, National Defense Medical Center, Taipei City, Taiwan
| |
Collapse
|
48
|
Lin Y, Huang J, Zhou M, Pan C, Shan A, Zhou C. Association of serum klotho levels with different-staged vascular calcification status in patients with maintenance hemodialysis. BMC Nephrol 2022; 23:374. [DOI: 10.1186/s12882-022-02995-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/01/2022] [Indexed: 11/20/2022] Open
Abstract
Abstract
Background
Vascular calcification (VC) is suggested to be associated with serum klotho levels in patients with maintenance hemodialysis (MHD), whereas there is a lack of reports on the associations of VC status in whole arteries with serum klotho contents.
Methods
One hundred forty eligible patients with MHD and a total of age-and gender-matched normal controls (NCs) were recruited. We analyzed the VC statuses of large arteries and peripheral muscular arteries by calculating the sum of scores from each artery. The levels of serum klotho were determined by ELISA. In addition, the relationship between serum klotho and VC status was evaluated using correlation analysis and regression analysis.
Results
The VC severity in MHD patients tended to be worse in comparison with NCs. Serum klotho level in patients with MHD was lower than that in the NC subjects (P < 0.0001), which was correlated with VC scores as reflected by correlation analysis and regression analysis. Serum klotho concentrations exhibited a dynamic decline along with increased VC status stages. Subjects with higher levels of serum klotho had a higher prevalence of cardiovascular events.
Conclusion
Our study indicates serum klotho is strongly associated with VC status in a stage-dependent manner.
Collapse
|
49
|
Lisowska KA, Storoniak H, Soroczyńska-Cybula M, Maziewski M, Dębska-Ślizień A. Serum Levels of α-Klotho, Inflammation-Related Cytokines, and Mortality in Hemodialysis Patients. J Clin Med 2022; 11:6518. [PMID: 36362746 PMCID: PMC9656457 DOI: 10.3390/jcm11216518] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 08/18/2023] Open
Abstract
It has been hypothesized that α-Klotho deficiency might contribute to chronic inflammation in patients with end-stage renal disease (ESRD), especially those on hemodialysis (HD). Serum Klotho levels by some authors are considered a potential predictor of cerebrovascular events. Therefore, we analyzed serum levels of α-Klotho with ELISA and inflammation-related cytokines in HD patients. Sixty-seven HD patients and 19 healthy people were recruited between November 2017 and June 2021. A Cytometric Bead Array (CBA) was used to determine the level of different cytokines: IL-12p70, TNF, IL-10, IL-6, IL-1β, and IL-8. A human Klotho ELISA kit was used to determine the level of α-Klotho in the plasma samples of HD patients. There was no difference in serum levels of α-Klotho between HD patients and healthy people. Patients had increased serum IL-6 and IL-8. Significant positive correlations existed between the concentration of α-Klotho and the serum concentrations of IL-12p70, IL-10, and IL-1β. However, in a multivariable linear regression analysis, only patients' age was associated independently with α-Klotho level. Serum α-Klotho was not associated with higher mortality risk in HD patients. While these results draw attention to potential relationships between α-Klotho proteins and inflammatory markers in HD patients, our cross-sectional study could not confirm the pathogenic link between α-Klotho, inflammation, and cardiovascular mortality.
Collapse
Affiliation(s)
| | - Hanna Storoniak
- Department of Nephrology, Transplantology and Internal Medicine, Faculty of Medicine, Medical University of Gdańsk, 80-214 Gdańsk, Poland
| | - Monika Soroczyńska-Cybula
- Department of Pathophysiology, Faculty of Medicine, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Mateusz Maziewski
- Department of Pathophysiology, Faculty of Medicine, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Alicja Dębska-Ślizień
- Department of Nephrology, Transplantology and Internal Medicine, Faculty of Medicine, Medical University of Gdańsk, 80-214 Gdańsk, Poland
| |
Collapse
|
50
|
Zhao Y, Zeng X, Xu X, Wang W, Xu L, Wu Y, Li H. Low-dose 5-aza-2'-deoxycytidine protects against early renal injury by increasing klotho expression. Epigenomics 2022; 14:1411-1425. [PMID: 36695107 DOI: 10.2217/epi-2022-0430] [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: 01/26/2023] Open
Abstract
Aim: To explore the effect of the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (Aza) on early renal injury. Materials & methods: Cell damage and inflammation are features of early renal injury. The apoptosis and inflammation in hypoxia/reoxygenation (H/R)-induced human proximal tubular epithelial cells (HK-2) and ischemia-reperfusion kidney were studied, and expression of the protein klotho was investigated. Results: Aza induced HK-2 apoptosis in a dose-dependent manner, but low-dose Aza attenuated the apoptosis and inflammation in H/R-induced HK-2 cells and ischemia-reperfusion kidney. Low-dose Aza ameliorated renal function in mice with renal ischemia-reperfusion injury. Meanwhile, low-dose Aza upregulated klotho expression in H/R-induced HK-2 cells and ischemia-reperfusion kidney. Klotho knockdown abrogated the effects of low-dose Aza on apoptosis and inflammation. Conclusion: Low-dose Aza protects against renal early injury by increasing klotho expression.
Collapse
Affiliation(s)
- Yanlong Zhao
- Dialysis Department of Nephrology Hospital, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi, 710003, China
| | - Xiaorong Zeng
- Dialysis Department of Nephrology Hospital, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi, 710003, China
| | - Xinli Xu
- Dialysis Department of Nephrology Hospital, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi, 710003, China
| | - Wenjing Wang
- Graduate School, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, China
| | - Lei Xu
- Graduate School, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, China
| | - Yiying Wu
- Graduate School, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, China
| | - Hang Li
- Graduate School, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, China
| |
Collapse
|