1
|
Javier AJS, Kennedy FM, Yi X, Wehling-Henricks M, Tidball JG, White KE, Witczak CA, Kuro-O M, Welc SS. Klotho Is Cardioprotective in the mdx Mouse Model of Duchenne Muscular Dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:923-940. [PMID: 39889824 PMCID: PMC12016860 DOI: 10.1016/j.ajpath.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/18/2024] [Accepted: 12/30/2024] [Indexed: 02/03/2025]
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, progressive skeletal and cardiac myopathy. Cardiomyopathy is the leading cause of death in patients with DMD, but the molecular basis for heart failure is incompletely understood. As with humans, in the mdx mouse model of DMD, cardiac function is impaired after the onset of skeletal muscle pathology. Dysregulation of Klotho gene regulation in dystrophic skeletal muscles occurs at disease onset, affecting pathogenesis. Whether Klotho is protective against dystrophin-deficient cardiomyopathy is unknown. This study found that expression of a Klotho transgene prevented deficits in left ventricular ejection fraction and fractional shortening in mdx mice. Improvements in cardiac performance were associated with reductions in adverse cardiac remodeling, cardiac myocyte hypertrophy, and fibrosis. In addition, mdx mice expressed high concentrations of plasma fibroblast growth factor 23 (FGF23), and expression was increased locally in hearts. The cardioprotective effects of Klotho were not associated with differences in renal function or serum biochemistries, but transgene expression prevented increased expression of plasma FGF23 and cardiac Fgf23 mRNA expression. Cardiac reactive oxygen species, oxidative damage, mitochondrial damage, and apoptosis were reduced in transgenic hearts. FGF23 stimulated hypertrophic growth in dystrophic neonatal mouse ventricular myocytes in vitro, which was inhibited by co-stimulation with soluble Klotho. Taken together, these results show that Klotho prevented dystrophic cardiac remodeling and improved function.
Collapse
MESH Headings
- Animals
- Klotho Proteins
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/physiopathology
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/complications
- Muscular Dystrophy, Duchenne/genetics
- Glucuronidase/metabolism
- Glucuronidase/genetics
- Fibroblast Growth Factor-23
- Fibroblast Growth Factors/metabolism
- Fibroblast Growth Factors/blood
- Fibroblast Growth Factors/genetics
- Mice, Inbred mdx
- Disease Models, Animal
- Mice
- Cardiomyopathies/pathology
- Cardiomyopathies/metabolism
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Mice, Transgenic
- Mice, Inbred C57BL
- Male
- Apoptosis
- Reactive Oxygen Species/metabolism
- Fibrosis
- Humans
- Myocardium/pathology
- Myocardium/metabolism
Collapse
Affiliation(s)
- Areli Jannes S Javier
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana
| | - Felicia M Kennedy
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Xin Yi
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - James G Tidball
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, California
| | - Kenneth E White
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Carol A Witczak
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Steven S Welc
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana.
| |
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
|
Williams MJ, Patel HM, Halling CB, Hruska KA. The Impact of a Western Diet High in Phosphate on the CKD-MBD in an Alport Syndrome Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633378. [PMID: 39896481 PMCID: PMC11785106 DOI: 10.1101/2025.01.17.633378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Background Chronic kidney disease - mineral bone disorder (CKD-MBD) is a syndrome that begins early in CKD, contributes to CKD-associated mortality, and includes components of FGF23 elevation, αklotho deficiency, CKD-stimulated vascular disease, and renal osteodystrophy. Hyperphosphatemia, occurring in later stages of CKD, is also driven by mechanisms of CKD-MBD, and has been shown to stimulate vascular calcification. In a mouse model of Alport CKD that is resistant to vascular calcification, we examine the effects of a high-phosphate Western-type diet on the CKD-MBD, and test whether the diet promotes induction of vascular calcification. Methods An X-linked Col4a5 deficient murine homolog of Alport Syndrome (CKD) and wild type (WT) littermates were fed an animal protein 1.2% high phosphate diet or a standard vegetable protein diet. At disease progression equivalent to CKD stage 4-5, we examined kidney histology for fibrosis, blood for BUN (marker of CKD), and markers of CKD-MBD disease progression, kidney tissue for klotho production, and aorta histology and tissue mRNA and protein analysis for vascular calcification. Results The Western high Pi diet produced hyperphosphatemia in the CKD animals compared to WT and increased plasma PTH (1880 from 110 pg / ml), FGF23 c-term (670 from 120 pg / ml), and FGF23 intact (3780 from 280 pg / ml), and reduced kidney klotho mRNA and protein (57-67% reduction) (all p < 0.01). Referenced against the CKD animals fed vegetable-based diet, the Western high phosphate-fed CKD animals showed higher levels of plasma PTH and FGF23s. In the wild-type control mice with normal renal function, Western diet produced increased PTH, intact FGF23, and reduced renal klotho (all p <0.01). Vascular smooth muscle transdifferentiation and vascular calcification was not induced by Western high phosphate diet in this model of CKD. Conclusions Our results show that a Western-style high-phosphate diet advances elements of the CKD-MBD. Renal klotho, FGF23 and PTH are affected by diet even with normal kidney function, suggesting a need for early intervention in the management of phosphate homeostasis as a component of CKD therapy. Additionally, CKD, klotho, and FGF23 all are associated with early aging. Therefore, our findings suggest that a Western high Pi diet accelerates aging and would contribute to the systemic complications of CKD - cardiac disease, osteodystrophy, and vascular disease.
Collapse
|
4
|
Wang S, Sun W, Ding C, Zhou W, Zhang M, Xu H. The role of serum α-Klotho levels in preventing hearing impairment among middle-aged and older adults: insights from a nationally representative sample. Front Aging Neurosci 2024; 16:1415494. [PMID: 39610715 PMCID: PMC11602472 DOI: 10.3389/fnagi.2024.1415494] [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: 04/10/2024] [Accepted: 10/24/2024] [Indexed: 11/30/2024] Open
Abstract
Background The Klotho gene is implicated in suppressing aging phenotypes and influencing age-related diseases. Previous studies have delved into its connection with different diseases, yet the association between Klotho and hearing loss has rarely been examined. A recent population study explored the relationship between serum Klotho and hearing loss, but it had certain limitations. This study aims to analyze the link between serum α-Klotho levels and hearing thresholds, as well as the risk of hearing loss. Methods A total of 1,762 adults aged 40-69 years were selected from the 2011-2012 National Health and Nutrition Examination Survey (NHANES). Data on audiometry, serum α-Klotho levels, and relevant covariates were gathered. Statistical analyses, including linear and logistic regression, assessed the relationships of serum α-Klotho levels with hearing outcomes. Results Increased serum α-Klotho levels were correlated with diminished hearing thresholds and a lower risk of hearing loss. Quartile analysis revealed a significant trend, where elevated α-Klotho levels were linked to better auditory outcomes. Adjusted models controlled for various covariates, affirming the robustness of the findings. Non-linear associations were not observed. Conclusion This study provided novel evidence of a negative association between serum α-Klotho and hearing impairment in adults aged 40-69. Our results suggested a protective role of serum α-Klotho on adults with hearing loss.
Collapse
Affiliation(s)
- Siyuan Wang
- School of Public Health, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wen Sun
- School of Public Health, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chan Ding
- School of Public Health, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wenxin Zhou
- School of Public Health, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Min Zhang
- School of Medical Humanities and Management (School of General Practice Competency Education), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Huadong Xu
- School of Public Health, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Edmonston D, Fuchs MAA, Burke EJ, Isakova T, Wolf M. Klotho and Clinical Outcomes in CKD: Findings From the Chronic Renal Insufficiency Cohort (CRIC) Study. Am J Kidney Dis 2024; 84:349-360.e1. [PMID: 38583756 PMCID: PMC11344676 DOI: 10.1053/j.ajkd.2024.02.008] [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: 06/28/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 04/09/2024]
Abstract
RATIONALE & OBJECTIVE Klotho deficiency may affect clinical outcomes in chronic kidney disease (CKD) through fibroblast growth factor-23 (FGF23)-dependent and -independent pathways. However, the association between circulating Klotho and clinical outcomes in CKD remains unresolved and was the focus of this study. STUDY DESIGN Prospective observational study. SETTING & PARTICIPANTS 1,088 participants in the Chronic Renal Insufficiency Cohort (CRIC) Study with an estimated glomerular filtration rate (eGFR) of 20-70mL/min/1.73m2. EXPOSURE Plasma Klotho level at the year-1 study visit. OUTCOMES 5-year risks of all-cause mortality, heart failure hospitalization, atherosclerotic cardiovascular events, and a composite kidney end point that comprised a sustained 50% decrease in eGFR, dialysis, kidney transplant, or eGFR<15mL/min/1.73m2. ANALYTICAL APPROACH We divided Klotho into 6 groups to account for its nonnormal distribution. We used Cox proportional hazards regression and subdistribution hazards models to compare survival and clinical outcomes, respectively, between Klotho groups. We sequentially adjusted for demographic characteristics, kidney function, cardiovascular risk factors, sample age, and FGF23. RESULTS Mean eGFR was 42mL/min/1.73m2, and median Klotho concentration was 0.31ng/mL (IQR, 0.10-3.27ng/mL). When compared with the lowest Klotho group, survival (HR, 0.77; 95% CI, 0.32-1.89), heart failure hospitalization (HR, 1.10; 95% CI, 0.38-3.17), atherosclerotic cardiovascular events (HR, 1.19; 95% CI, 0.57-2.52), and CKD progression (HR, 1.05; 95% CI, 0.58-1.91) did not differ in the high Klotho group. In contrast, FGF23 was significantly associated with mortality and heart failure hospitalization independent of Klotho levels. LIMITATIONS Despite adjustments, we cannot exclude the potential influence of residual confounding or sample storage on the results. A single measurement of plasma Klotho concentration may not capture Klotho patterns over time. CONCLUSIONS In a large, diverse, well-characterized CKD cohort, Klotho was not associated with clinical outcomes, and Klotho deficiency did not confound the association of FGF23 with mortality or heart failure hospitalization. PLAIN-LANGUAGE SUMMARY Klotho is a protein that is vital to mineral metabolism and aging and may protect against cardiovascular disease. Klotho levels decrease in chronic kidney disease (CKD), but the association between Klotho and clinical outcomes in CKD remains uncertain. In a prospective cohort study of more than 1,000 people with CKD, circulating Klotho levels were not associated with kidney disease progression, cardiovascular outcomes, or mortality. These results suggest that the decrease in circulating Klotho levels in CKD does not play a prominent role in the development of poor clinical outcomes.
Collapse
Affiliation(s)
- Daniel Edmonston
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC; Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC.
| | - Michaela A A Fuchs
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Emily J Burke
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Tamara Isakova
- Division of Nephrology and Hypertension, Department of Medicine and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Myles Wolf
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC; Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| |
Collapse
|
6
|
Prud’homme GJ, Wang Q. Anti-Inflammatory Role of the Klotho Protein and Relevance to Aging. Cells 2024; 13:1413. [PMID: 39272986 PMCID: PMC11394293 DOI: 10.3390/cells13171413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/17/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
The α-Klotho protein (hereafter Klotho) is an obligate coreceptor for fibroblast growth factor 23 (FGF23). It is produced in the kidneys, brain and other sites. Klotho insufficiency causes hyperphosphatemia and other anomalies. Importantly, it is associated with chronic pathologies (often age-related) that have an inflammatory component. This includes atherosclerosis, diabetes and Alzheimer's disease. Its mode of action in these diseases is not well understood, but it inhibits or regulates multiple major pathways. Klotho has a membrane form and a soluble form (s-Klotho). Cytosolic Klotho is postulated but not well characterized. s-Klotho has endocrine properties that are incompletely elucidated. It binds to the FGF receptor 1c (FGFR1c) that is widely expressed (including endothelial cells). It also attaches to soluble FGF23, and FGF23/Klotho binds to FGFRs. Thus, s-Klotho might be a roaming FGF23 coreceptor, but it has other functions. Notably, Klotho (cell-bound or soluble) counteracts inflammation and appears to mitigate related aging (inflammaging). It inhibits NF-κB and the NLRP3 inflammasome. This inflammasome requires priming by NF-κB and produces active IL-1β, membrane pores and cell death (pyroptosis). In accord, Klotho countered inflammation and cell injury induced by toxins, damage-associated molecular patterns (DAMPs), cytokines, and reactive oxygen species (ROS). s-Klotho also blocks the TGF-β receptor and Wnt ligands, which lessens fibrotic disease. Low Klotho is associated with loss of muscle mass (sarcopenia), as occurs in aging and chronic diseases. s-Klotho counters the inhibitory effects of myostatin and TGF-β on muscle, reduces inflammation, and improves muscle repair following injury. The inhibition of TGF-β and other factors may also be protective in diabetic retinopathy and age-related macular degeneration (AMD). This review examines Klotho functions especially as related to inflammation and potential applications.
Collapse
Affiliation(s)
- Gérald J. Prud’homme
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 220 Walmer Rd, Toronto, ON M5R 3R7, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON M5B 1W8, Canada
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai 200030, China
- Shanghai Innogen Pharmaceutical Co., Ltd., Shanghai 201318, China
| |
Collapse
|
7
|
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
|
8
|
Kozłowski K, Konończuk K, Muszyńska-Rosłan K, Żelazowska-Rutkowska B, Taranta-Janusz K, Werbel K, Krawczuk-Rybak M, Latoch E. Circulating Levels of Soluble α-Klotho and FGF23 in Childhood Cancer Survivors: Lack of Association with Nephro- and Cardiotoxicity-A Preliminary Study. J Clin Med 2024; 13:2968. [PMID: 38792509 PMCID: PMC11122186 DOI: 10.3390/jcm13102968] [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: 04/22/2024] [Revised: 05/05/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
Background/Objectives: The survival rate among pediatric cancer patients has reached 80%; however, these childhood cancer survivors (CCSs) are at a heightened risk of developing chronic conditions in adulthood, particularly kidney and cardiovascular diseases. The aims of this study were to assess the serum α-Klotho and FGF23 levels in CCSs and to determine their association with nephro- and cardiotoxicity. Methods: This study evaluated a cohort of 66 CCSs who remained in continuous remission, with a mean follow-up of 8.41 ± 3.76 years. Results: The results of this study revealed that CCSs exhibited significantly higher levels of soluble α-Klotho compared to healthy peers (1331.4 ± 735.5 pg/mL vs. 566.43 ± 157.7 pg/mL, p < 0.0001), while no significant difference was observed in their FGF23 levels. Within the participant cohort, eight individuals (12%) demonstrated a reduced estimated glomerular filtration rate (eGFR) below 90 mL/min/1.73 m2. The relationship between treatment with abdominal radiotherapy and reduced eGFR was confirmed (p < 0.05). No correlations were found between potential treatment-related risk factors, such as chemotherapy or radiation therapy, serum levels of α-Klotho and FGF23, and nephro- and cardiotoxicity. Conclusions: In conclusion, this preliminary cross-sectional study revealed elevated levels of α-Klotho among childhood cancer survivors but did not establish a direct association with anticancer treatment. The significance of elevated α-Klotho protein levels among CCSs warrants further investigation.
Collapse
Affiliation(s)
- Kacper Kozłowski
- Department of Pediatric Oncology and Hematology, Medical University of Bialystok, 15-274 Białystok, Poland; (K.K.); (K.K.); (K.M.-R.); (M.K.-R.)
| | - Katarzyna Konończuk
- Department of Pediatric Oncology and Hematology, Medical University of Bialystok, 15-274 Białystok, Poland; (K.K.); (K.K.); (K.M.-R.); (M.K.-R.)
| | - Katarzyna Muszyńska-Rosłan
- Department of Pediatric Oncology and Hematology, Medical University of Bialystok, 15-274 Białystok, Poland; (K.K.); (K.K.); (K.M.-R.); (M.K.-R.)
| | - Beata Żelazowska-Rutkowska
- Department of Pediatric Laboratory Diagnostics, Medical University of Bialystok, 15-274 Białystok, Poland;
| | - Katarzyna Taranta-Janusz
- Department of Pediatrics and Nephrology, Medical University of Bialystok, 15-274 Białystok, Poland;
| | - Katarzyna Werbel
- Department of Neonatology, Pathology and Newborn Intensive Care, Jędrzej Śniadecki Independent Public Healthcare Center—Regional Hospital, 15-027 Białystok, Poland;
| | - Maryna Krawczuk-Rybak
- Department of Pediatric Oncology and Hematology, Medical University of Bialystok, 15-274 Białystok, Poland; (K.K.); (K.K.); (K.M.-R.); (M.K.-R.)
| | - Eryk Latoch
- Department of Pediatric Oncology and Hematology, Medical University of Bialystok, 15-274 Białystok, Poland; (K.K.); (K.K.); (K.M.-R.); (M.K.-R.)
| |
Collapse
|
9
|
Williams MJ, Halabi CM, Patel HM, Joseph Z, McCommis K, Weinheimer C, Kovacs A, Lima F, Finck B, Malluche H, Hruska KA. In chronic kidney disease altered cardiac metabolism precedes cardiac hypertrophy. Am J Physiol Renal Physiol 2024; 326:F751-F767. [PMID: 38385175 PMCID: PMC11386984 DOI: 10.1152/ajprenal.00416.2023] [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: 12/22/2023] [Revised: 02/15/2024] [Accepted: 02/15/2024] [Indexed: 02/23/2024] Open
Abstract
Conduit arterial disease in chronic kidney disease (CKD) is an important cause of cardiac complications. Cardiac function in CKD has not been studied in the absence of arterial disease. In an Alport syndrome model bred not to have conduit arterial disease, mice at 225 days of life (dol) had CKD equivalent to humans with CKD stage 4-5. Parathyroid hormone (PTH) and FGF23 levels were one log order elevated, circulating sclerostin was elevated, and renal activin A was strongly induced. Aortic Ca levels were not increased, and vascular smooth muscle cell (VSMC) transdifferentiation was absent. The CKD mice were not hypertensive, and cardiac hypertrophy was absent. Freshly excised cardiac tissue respirometry (Oroboros) showed that ADP-stimulated O2 flux was diminished from 52 to 22 pmol/mg (P = 0.022). RNA-Seq of cardiac tissue from CKD mice revealed significantly decreased levels of cardiac mitochondrial oxidative phosphorylation genes. To examine the effect of activin A signaling, some Alport mice were treated with a monoclonal Ab to activin A or an isotype-matched IgG beginning at 75 days of life until euthanasia. Treatment with the activin A antibody (Ab) did not affect cardiac oxidative phosphorylation. However, the activin A antibody was active in the skeleton, disrupting the effect of CKD to stimulate osteoclast number, eroded surfaces, and the stimulation of osteoclast-driven remodeling. The data reported here show that cardiac mitochondrial respiration is impaired in CKD in the absence of conduit arterial disease. This is the first report of the direct effect of CKD on cardiac respiration.NEW & NOTEWORTHY Heart disease is an important morbidity of chronic kidney disease (CKD). Hypertension, vascular stiffness, and vascular calcification all contribute to cardiac pathophysiology. However, cardiac function in CKD devoid of vascular disease has not been studied. Here, in an animal model of human CKD without conduit arterial disease, we analyze cardiac respiration and discover that CKD directly impairs cardiac mitochondrial function by decreasing oxidative phosphorylation. Protection of cardiac oxidative phosphorylation may be a therapeutic target in CKD.
Collapse
Affiliation(s)
- Matthew J Williams
- Renal Division, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Carmen M Halabi
- Renal Division, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Hiral M Patel
- Renal Division, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Zachary Joseph
- Renal Division, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Kyle McCommis
- Geriatrics and Nutritional Science Division, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Carla Weinheimer
- Cardiology Division, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Attila Kovacs
- Cardiology Division, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Florence Lima
- Renal Division, Department of Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - Brian Finck
- Geriatrics and Nutritional Science Division, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Hartmut Malluche
- Renal Division, Department of Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - Keith A Hruska
- Renal Division, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, United States
- Renal Division, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States
- Department of Cell Biology, Washington University in St. Louis, St. Louis, Missouri, United States
| |
Collapse
|
10
|
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
|
11
|
Hu MC, Reneau JA, Shi M, Takahashi M, Chen G, Mohammadi M, Moe OW. C-terminal fragment of fibroblast growth factor 23 improves heart function in murine models of high intact fibroblast growth factor 23. Am J Physiol Renal Physiol 2024; 326:F584-F599. [PMID: 38299214 PMCID: PMC11208029 DOI: 10.1152/ajprenal.00298.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 02/02/2024] Open
Abstract
Cardiovascular disease (CVD) is the major cause of death in chronic kidney disease (CKD) and is associated with high circulating fibroblast growth factor (FGF)23 levels. It is unresolved whether high circulating FGF23 is a mere biomarker or pathogenically contributes to cardiomyopathy. It is also unknown whether the C-terminal FGF23 peptide (cFGF23), a natural FGF23 antagonist proteolyzed from intact FGF23 (iFGF23), retards CKD progression and improves cardiomyopathy. We addressed these questions in three murine models with high endogenous FGF23 and cardiomyopathy. First, we examined wild-type (WT) mice with CKD induced by unilateral ischemia-reperfusion and contralateral nephrectomy followed by a high-phosphate diet. These mice were continuously treated with intraperitoneal implanted osmotic minipumps containing either iFGF23 protein to further escalate FGF23 bioactivity, cFGF23 peptide to block FGF23 signaling, vehicle, or scrambled peptide as negative controls. Exogenous iFGF23 protein given to CKD mice exacerbated pathological cardiac remodeling and CKD progression, whereas cFGF23 treatment improved heart and kidney function, attenuated fibrosis, and increased circulating soluble Klotho. WT mice without renal insult placed on a high-phosphate diet and homozygous Klotho hypomorphic mice, both of whom develop moderate CKD and clear cardiomyopathy, were treated with cFGF23 or vehicle. Mice treated with cFGF23 in both models had improved heart and kidney function and histopathology. Taken together, these data indicate high endogenous iFGF23 is not just a mere biomarker but pathogenically deleterious in CKD and cardiomyopathy. Furthermore, attenuation of FGF23 bioactivity by cFGF23 peptide is a promising therapeutic strategy to protect the kidney and heart from high FGF23 activity.NEW & NOTEWORTHY There is a strong correlation between cardiovascular morbidity and high circulating fibroblast growth factor 23 (FGF23) levels, but causality was never proven. We used a murine chronic kidney disease (CKD) model to show that intact FGF23 (iFGF23) is pathogenic and contributes to both CKD progression and cardiomyopathy. Blockade of FGF23 signaling with a natural proteolytic product of iFGF23, C-terminal FGF23, alleviated kidney and cardiac histology, and function in three separate murine models of high endogenous FGF23.
Collapse
Affiliation(s)
- Ming Chang Hu
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - James A Reneau
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Mingjun Shi
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Masaya Takahashi
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, United States
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Gaozhi Chen
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Moosa Mohammadi
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Orson W Moe
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, United States
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
12
|
Martín-Vírgala J, Martín-Carro B, Fernández-Villabrille S, Ruiz-Torres MP, Gómez-Alonso C, Rodríguez-García M, Fernández-Martín JL, Alonso-Montes C, Panizo S, Cannata-Andía JB, Naves-Díaz M, Carrillo-López N. Soluble Klotho, a Potential Biomarker of Chronic Kidney Disease-Mineral Bone Disorders Involved in Healthy Ageing: Lights and Shadows. Int J Mol Sci 2024; 25:1843. [PMID: 38339121 PMCID: PMC10855561 DOI: 10.3390/ijms25031843] [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: 12/28/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Shortly after the discovery of Klotho, interest grew in its potential role in chronic kidney disease (CKD). There are three isoforms of the Klotho protein: αKlotho, βKlotho and γKlotho. This review will focus on αKlotho due to its relevance as a biomarker in CKD. αKlotho is synthesized mainly in the kidneys, but it can be released into the bloodstream and urine as soluble Klotho (sKlotho), which undertakes systemic actions, independently or in combination with FGF23. It is usually accepted that sKlotho levels are reduced early in CKD and that lower levels of sKlotho might be associated with the main chronic kidney disease-mineral bone disorders (CKD-MBDs): cardiovascular and bone disease. However, as results are inconsistent, the applicability of sKlotho as a CKD-MBD biomarker is still a matter of controversy. Much of the inconsistency can be explained due to low sample numbers, the low quality of clinical studies, the lack of standardized assays to assess sKlotho and a lack of consensus on sample processing, especially in urine. In recent decades, because of our longer life expectancies, the prevalence of accelerated-ageing diseases, such as CKD, has increased. Exercise, social interaction and caloric restriction are considered key factors for healthy ageing. While exercise and social interaction seem to be related to higher serum sKlotho levels, it is not clear whether serum sKlotho might be influenced by caloric restriction. This review focuses on the possible role of sKlotho as a biomarker in CKD-MBD, highlighting the difference between solid knowledge and areas requiring further research, including the role of sKlotho in healthy ageing.
Collapse
Affiliation(s)
- Julia Martín-Vírgala
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain;
| | - Beatriz Martín-Carro
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain;
| | - Sara Fernández-Villabrille
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain;
| | - María Piedad Ruiz-Torres
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain;
- Área 5—Fisiología y Fisiopatología Renal y Vascular del Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Physiology Unit, Department of Systems Biology, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá, 28871 Alcalá de Henares, Spain
| | - Carlos Gómez-Alonso
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Minerva Rodríguez-García
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain;
- Nephrology Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - José Luis Fernández-Martín
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain;
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Cristina Alonso-Montes
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain;
| | - Sara Panizo
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain;
| | - Jorge B. Cannata-Andía
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain;
- Department of Medicine, Universidad de Oviedo, 33011 Oviedo, Spain
| | - Manuel Naves-Díaz
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain;
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Natalia Carrillo-López
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS2040, Kidney Disease), 28040 Madrid, Spain;
| |
Collapse
|
13
|
Edmonston D, Grabner A, Wolf M. FGF23 and klotho at the intersection of kidney and cardiovascular disease. Nat Rev Cardiol 2024; 21:11-24. [PMID: 37443358 DOI: 10.1038/s41569-023-00903-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/13/2023] [Indexed: 07/15/2023]
Abstract
Cardiovascular disease is the leading cause of death in patients with chronic kidney disease (CKD). As CKD progresses, CKD-specific risk factors, such as disordered mineral homeostasis, amplify traditional cardiovascular risk factors. Fibroblast growth factor 23 (FGF23) regulates mineral homeostasis by activating complexes of FGF receptors and transmembrane klotho co-receptors. A soluble form of klotho also acts as a 'portable' FGF23 co-receptor in tissues that do not express klotho. In progressive CKD, rising circulating FGF23 levels in combination with decreasing kidney expression of klotho results in klotho-independent effects of FGF23 on the heart that promote left ventricular hypertrophy, heart failure, atrial fibrillation and death. Emerging data suggest that soluble klotho might mitigate some of these effects via several candidate mechanisms. More research is needed to investigate FGF23 excess and klotho deficiency in specific cardiovascular complications of CKD, but the pathophysiological primacy of FGF23 excess versus klotho deficiency might never be precisely resolved, given the entangled feedback loops that they share. Therefore, randomized trials should prioritize clinical practicality over scientific certainty by targeting disordered mineral homeostasis holistically in an effort to improve cardiovascular outcomes in patients with CKD.
Collapse
Affiliation(s)
- Daniel Edmonston
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Alexander Grabner
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Myles Wolf
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
14
|
Arroyo E, Leber CA, Burney HN, Narayanan G, Moorthi R, Avin KG, Warden SJ, Moe SM, Lim K. Relationship between klotho and physical function in healthy aging. Sci Rep 2023; 13:21158. [PMID: 38036596 PMCID: PMC10689840 DOI: 10.1038/s41598-023-47791-5] [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: 03/21/2023] [Accepted: 11/18/2023] [Indexed: 12/02/2023] Open
Abstract
Epidemiological studies have reported a strong association between circulating Klotho and physical function; however, the cohorts were comprised of older adults with multiple comorbidities. Herein, we examined the relationship between Klotho and physical function in a community-based cohort of healthy adults. In this cross-sectional study, serum Klotho was measured in 80 adults who visited the Musculoskeletal Function, Imaging, and Tissue Resource Core of the Indiana Center for Musculoskeletal Health. Participants (n = 20, 10 [50%] men per group) were chosen into four age groups: 20-34, 35-49, 50-64, and ≥ 65 years, and were further grouped based on performance (low vs. high) on grip strength and chair stand tests. Klotho levels were lower in the ≥ 65 years group (703.0 [189.3] pg/mL; p = 0.022) and the 50-64 years group (722.6 [190.5] pg/mL; p = 0.045) compared to 20-34 years (916.1 [284.8] pg/mL). No differences were observed in Klotho between the low and high performers. The ≥ 65 years group walked a shorter distance during the 6-min walk test (6MWT) compared to 20-34 years (p = 0.005). Klotho was correlated with age (p < 0.001), body fat (p = 0.037), and 6MWT distance (p = 0.022). Klotho levels decline as early as the fifth decade of life, potentially before the onset of age-related impairment in exercise capacity.
Collapse
Affiliation(s)
- Eliott Arroyo
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Health and Exercise Science, Wake Forest University, Winston-Salem, NC, USA
| | - Cecilia A Leber
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Heather N Burney
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Gayatri Narayanan
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ranjani Moorthi
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Keith G Avin
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Physical Therapy, Indiana University School of Health and Human Sciences, Indianapolis, IN, USA
| | - Stuart J Warden
- Department of Physical Therapy, Indiana University School of Health and Human Sciences, Indianapolis, IN, USA
| | - Sharon M Moe
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kenneth Lim
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
15
|
Neyra JA, Moe OW. Bone Dysregulation in Acute Kidney Injury. Nephron Clin Pract 2023; 147:747-753. [PMID: 37757785 DOI: 10.1159/000534228] [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: 07/03/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Acute kidney injury (AKI) is a highly prevalent condition with multiple acute and chronic consequences. Survivors of AKI are at risk of AKI-to-chronic kidney disease (CKD) transition, which carries significant morbidity and mortality. One retrospective analysis showed increased risk of bone fracture post-AKI in humans, which was independent of CKD development. While there are several theoretical reasons for late disturbances of bone health post-AKI, no definitive data are available to date. An important question is whether there are bone sequelae from AKI that are independent of CKD, meaning bone disease prior to the onset, or in the absence of CKD - a form of "post-AKI osteopathy." While preclinical studies examining bone health after acute stressors have focused mostly on sepsis models, multiple experimental AKI models are readily available for longitudinal bone health interrogation. Future research should be tailored to define whether AKI is a risk factor, independent of CKD, for bone disease and if present, the time course and type of bone disease. This review summarizes a fraction of the existing data to provide some guidance in future research efforts.
Collapse
Affiliation(s)
- Javier A Neyra
- Department of Medicine, University of Alabama, Birmingham, Alabama, USA
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Departments of Internal Medicine and Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
16
|
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
|
17
|
Serum and Urinary Soluble α-Klotho as Markers of Kidney and Vascular Impairment. Nutrients 2023; 15:nu15061470. [PMID: 36986200 PMCID: PMC10057552 DOI: 10.3390/nu15061470] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/12/2023] [Accepted: 03/16/2023] [Indexed: 03/22/2023] Open
Abstract
This study was designed to investigate the controversy on the potential role of sKlotho as an early biomarker in Chronic Kidney Disease–Mineral Bone Disorder (CKD-MBD), to assess whether sKlotho is a reliable marker of kidney α-Klotho, to deepen the effects of sKlotho on vascular smooth muscle cells (VSMCs) osteogenic differentiation and to evaluate the role of autophagy in this process. Experimental studies were conducted in CKD mice fed a normal phosphorus (CKD+NP) or high phosphorus (CKD+HP) diet for 14 weeks. The patients’ study was performed in CKD stages 2–5 and in vitro studies which used VSMCs exposed to non-calcifying medium or calcifying medium with or without sKlotho. The CKD experimental model showed that the CKD+HP group reached the highest serum PTH, P and FGF23 levels, but the lowest serum and urinary sKlotho levels. In addition, a positive correlation between serum sKlotho and kidney α-Klotho was found. CKD mice showed aortic osteogenic differentiation, together with increased autophagy. The human CKD study showed that the decline in serum sKlotho is previous to the rise in FGF23. In addition, both serum sKlotho and FGF23 levels correlated with kidney function. Finally, in VSMCs, the addition of sKlotho prevented osteogenic differentiation and induced autophagy. It can be concluded that serum sKlotho was the earliest CKD-MBD biomarker, a reliable indicator of kidney α-Klotho and that might protect against osteogenic differentiation by increasing autophagy. Nevertheless, further studies are needed to investigate the mechanisms of this possible protective effect.
Collapse
|
18
|
Donate-Correa J, Matos-Perdomo E, González-Luis A, Martín-Olivera A, Ortiz A, Mora-Fernández C, Navarro-González JF. The Value of Klotho in Kidney Transplantation. Transplantation 2023; 107:616-627. [PMID: 36253904 DOI: 10.1097/tp.0000000000004331] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Kidney transplant recipients have better survival rates and improved quality of life than long-term dialysis patients. However, delayed graft function, immunosuppressive therapy nephrotoxicity, and rejection episodes may compromise graft and patient survival. The KL gene is highly expressed in kidney tubular cells and encodes the antiaging and kidney-protective protein Klotho, which has membrane-anchored and soluble forms and regulates mineral metabolism. Klotho expression decreases during acute kidney injury or chronic kidney disease, and human chronic kidney disease shares features of accelerated aging with murine Klotho deficiency. In this work, we review clinical studies on the relationship between Klotho and kidney transplantation. Specifically, we address the dynamics of serum and kidney Klotho levels in donors and kidney transplant recipients, the role of Klotho as a marker of current graft function and graft outcomes, and the potential impact of Klotho on kidney protection in the transplantation context. A better understanding of the potential biomarker and therapeutic utility of Klotho in kidney transplant recipients may provide new insights into the control of graft function and new therapeutic strategies to preserve allograft function.
Collapse
Affiliation(s)
- Javier Donate-Correa
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, Santander, Spain
- Instituto de Tecnologías Biomédicas, University of La Laguna, Santa Cruz de Tenerife, Spain
| | - Emiliano Matos-Perdomo
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Doctoral and Graduate School, University of La Laguna, San Cristóbal de La Laguna, Tenerife, Spain
| | - Ainhoa González-Luis
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Instituto de Tecnologías Biomédicas, University of La Laguna, Santa Cruz de Tenerife, Spain
- Doctoral and Graduate School, University of La Laguna, San Cristóbal de La Laguna, Tenerife, Spain
| | - Alberto Martín-Olivera
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Instituto de Tecnologías Biomédicas, University of La Laguna, Santa Cruz de Tenerife, Spain
- Doctoral and Graduate School, University of La Laguna, San Cristóbal de La Laguna, Tenerife, Spain
| | - Alberto Ortiz
- Instituto de Investigación Sanitaria Fundación Jiménez-Díaz-Universidad Autónoma de Madrid, Madrid, Spain
- RICORS2040 (Red de Investigación Renal-RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Mora-Fernández
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, Santander, Spain
- RICORS2040 (Red de Investigación Renal-RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan F Navarro-González
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, Santander, Spain
- Instituto de Tecnologías Biomédicas, University of La Laguna, Santa Cruz de Tenerife, Spain
- RICORS2040 (Red de Investigación Renal-RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
19
|
Promising novel therapeutic targets for kidney disease: Emphasis on kidney-specific proteins. Drug Discov Today 2023; 28:103466. [PMID: 36509391 DOI: 10.1016/j.drudis.2022.103466] [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: 09/07/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Abstract
Worldwide, around 850 million people are diagnosed with kidney disease but the available treatment options are still limited. Preclinical studies propose a plethora of druggable targets that can attenuate kidney disease and could qualify as novel therapeutic strategies, although most of these targets still await clinical testing. Here, we review some promising candidate targets for chronic kidney disease: intermedin, periostin, sirtuin, the cannabinoid receptor, Klotho, and uromodulin. For acute kidney injury, we discuss Apelin, Elabela, growth differentiation factor-15, Fyn kinase, and Klotho. Target selection for further clinical development should consider redundancies with the standard of care, potential synergistic effects with existing treatments, as well as the potential of additional effects on the cardiovascular system as a common comorbidity in patients with kidney disease.
Collapse
|
20
|
Kim D, Lee S, Choi JY, Lee J, Lee HJ, Min JY, Min KB. Association of α-klotho and lead and cadmium: A cross-sectional study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 843:156938. [PMID: 35753483 DOI: 10.1016/j.scitotenv.2022.156938] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 04/17/2022] [Accepted: 06/20/2022] [Indexed: 06/15/2023]
Abstract
Epigenetic aging is associated with harmful health effects such as oxidative stress from heavy metal exposure. We considered the relationship between genes and heavy metals in association with oxidative stress and then investigated the association between serum α- klotho and lead and cadmium exposure among adults in the United States from 2007 to 2016 participating in the National Health and Nutrition Examination Survey (NHANES). Samples included 9800 adults aged 40 to 79 years with measurements of serum α-klotho, lead and cadmium, and complete covariate data. Lead and cadmium levels were measured by inductively coupled plasma mass spectrometry and serum α-klotho levels were measured using enzyme-linked immunosorbent assay (ELISA). Multivariate linear regression analysis was used to estimate the association between serum α-klotho and blood lead, blood cadmium, and urinary cadmium. A percent increase in blood lead, blood cadmium, and urinary cadmium was associated with a statistically significant 4.0 % (p < 0.001), 2.0 %, (p = 0.003) and 1.0 % (p = 0.020) decrease in serum klotho. After adjustment, a percent increase in blood lead was associated with a statistically significant 4.0 % (p < 0.001) decrease in serum klotho; blood and urinary cadmium did not show any statistically significant associations after adjustment (β (95 % CI), p-value for blood cadmium: 0.00 (-0.02-0.01), p = 0.573; urinary cadmium: -0.01 (-0.03-0.01), p = 0.210). Mean serum klotho levels showed a statistically significant decreasing trend with increasing blood lead quartiles (unadjusted and all-adjusted geometric means and 95 % confidence intervals of serum klotho (in pg/mL) for Q1, Q2, Q3, and Q4: unadjusted: 827.49 (814.20-840.92), 811.92 (794.73-829.48), 791.48 (775.11-808.19), and 772.01 (754.23-790.29); adjusted: 830.64 (805.53-856.45), 816.07 (789.18-843.87), 800.71 (773.71-828.57), and 784.31 (757.94-811.59)). Blood lead and levels were negatively associated with serum α-klotho levels in a representative population of US adults. These results suggest that blood lead levels may be associated with the serum levels of a protein associated with cognition and aging. Further research is recommended to investigate the causality behind such relationship.
Collapse
Affiliation(s)
- Donghoon Kim
- Department of Preventive Medicine, College of Medicine, Seoul National University,103 Daehak-ro, Jongno gu, Seoul 110-799, South Korea
| | - Sohyae Lee
- Department of Preventive Medicine, College of Medicine, Seoul National University,103 Daehak-ro, Jongno gu, Seoul 110-799, South Korea; Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea
| | - Ju-Young Choi
- Department of Preventive Medicine, College of Medicine, Seoul National University,103 Daehak-ro, Jongno gu, Seoul 110-799, South Korea
| | - Jaeho Lee
- Department of Preventive Medicine, College of Medicine, Seoul National University,103 Daehak-ro, Jongno gu, Seoul 110-799, South Korea; Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea
| | - Hyo-Jung Lee
- Department of Preventive Medicine, College of Medicine, Seoul National University,103 Daehak-ro, Jongno gu, Seoul 110-799, South Korea
| | - Jin-Young Min
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, South Korea.
| | - Kyoung-Bok Min
- Department of Preventive Medicine, College of Medicine, Seoul National University,103 Daehak-ro, Jongno gu, Seoul 110-799, South Korea; Institute of Health Policy and Management, Medical Research Center, Seoul National University, South Korea.
| |
Collapse
|
21
|
Yu LX, Li SS, Sha MY, Kong JW, Ye JM, Liu QF. The controversy of klotho as a potential biomarker in chronic kidney disease. Front Pharmacol 2022; 13:931746. [PMID: 36210812 PMCID: PMC9532967 DOI: 10.3389/fphar.2022.931746] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Klotho is an identified longevity gene with beneficial pleiotropic effects on the kidney. Evidence shows that a decline in serum Klotho level occurs in early chronic kidney disease (CKD) and continues as CKD progresses. Klotho deficiency is associated with poor clinical outcomes and CKD mineral bone disorders (CKD-MBD). Klotho has been postulated as a candidate biomarker in the evaluation of CKD. However, the evidence for the clinical significance of the relationship between Klotho and kidney function, CKD stage, adverse kidney and/or non-kidney outcomes, and CKD-MBD remains inconsistent and in some areas, contradictory. Therefore, there is uncertainty as to whether Klotho is a potential biomarker in CKD; a general consensus regarding the clinical significance of Klotho in CKD has not been reached, and there is limited evidence synthesis in this area. To address this, we have systematically assessed the areas of controversy, focusing on the inconsistencies in the evidence base. We used a PICOM strategy to search for relevant studies and the Newcastle–Ottawa Scale scoring to evaluate included publications. We reviewed the inconsistent clinical findings based on the relationship of Klotho with CKD stage, kidney and/or non-kidney adverse outcomes, and CKD-MBD in human studies. Subsequently, we assessed the underlying sources of the controversies and highlighted future directions to resolve these inconsistencies and clarify whether Klotho has a role as a biomarker in clinical practice in CKD.
Collapse
Affiliation(s)
- Li-Xia Yu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Sha-Sha Li
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Min-Yue Sha
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Jia-Wei Kong
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Jian-Ming Ye
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- *Correspondence: Jian-Ming Ye, ; Qi-Feng Liu,
| | - Qi-Feng Liu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- *Correspondence: Jian-Ming Ye, ; Qi-Feng Liu,
| |
Collapse
|
22
|
Prud’homme GJ, Kurt M, Wang Q. Pathobiology of the Klotho Antiaging Protein and Therapeutic Considerations. FRONTIERS IN AGING 2022; 3:931331. [PMID: 35903083 PMCID: PMC9314780 DOI: 10.3389/fragi.2022.931331] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/06/2022] [Indexed: 12/06/2022]
Abstract
The α-Klotho protein (henceforth denoted Klotho) has antiaging properties, as first observed in mice homozygous for a hypomorphic Klotho gene (kl/kl). These mice have a shortened lifespan, stunted growth, renal disease, hyperphosphatemia, hypercalcemia, vascular calcification, cardiac hypertrophy, hypertension, pulmonary disease, cognitive impairment, multi-organ atrophy and fibrosis. Overexpression of Klotho has opposite effects, extending lifespan. In humans, Klotho levels decline with age, chronic kidney disease, diabetes, Alzheimer’s disease and other conditions. Low Klotho levels correlate with an increase in the death rate from all causes. Klotho acts either as an obligate coreceptor for fibroblast growth factor 23 (FGF23), or as a soluble pleiotropic endocrine hormone (s-Klotho). It is mainly produced in the kidneys, but also in the brain, pancreas and other tissues. On renal tubular-cell membranes, it associates with FGF receptors to bind FGF23. Produced in bones, FGF23 regulates renal excretion of phosphate (phosphaturic effect) and vitamin D metabolism. Lack of Klotho or FGF23 results in hyperphosphatemia and hypervitaminosis D. With age, human renal function often deteriorates, lowering Klotho levels. This appears to promote age-related pathology. Remarkably, Klotho inhibits four pathways that have been linked to aging in various ways: Transforming growth factor β (TGF-β), insulin-like growth factor 1 (IGF-1), Wnt and NF-κB. These can induce cellular senescence, apoptosis, inflammation, immune dysfunction, fibrosis and neoplasia. Furthermore, Klotho increases cell-protective antioxidant enzymes through Nrf2 and FoxO. In accord, preclinical Klotho therapy ameliorated renal, cardiovascular, diabetes-related and neurodegenerative diseases, as well as cancer. s-Klotho protein injection was effective, but requires further investigation. Several drugs enhance circulating Klotho levels, and some cross the blood-brain barrier to potentially act in the brain. In clinical trials, increased Klotho was noted with renin-angiotensin system inhibitors (losartan, valsartan), a statin (fluvastatin), mTOR inhibitors (rapamycin, everolimus), vitamin D and pentoxifylline. In preclinical work, antidiabetic drugs (metformin, GLP-1-based, GABA, PPAR-γ agonists) also enhanced Klotho. Several traditional medicines and/or nutraceuticals increased Klotho in rodents, including astaxanthin, curcumin, ginseng, ligustilide and resveratrol. Notably, exercise and sport activity increased Klotho. This review addresses molecular, physiological and therapeutic aspects of Klotho.
Collapse
Affiliation(s)
- Gérald J. Prud’homme
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada
- *Correspondence: Gérald J. Prud’homme,
| | - Mervé Kurt
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China
- Shanghai Yinuo Pharmaceutical Co., Ltd., Shanghai, China
| |
Collapse
|
23
|
Sharma S, Katz R, Ginsberg C, Bullen A, Vallon V, Thomson S, Moe OW, Hoofnagle AN, de Leeuw PW, Kroon AA, Houben AJHM, Ix JH. Renal Clearance of Fibroblast Growth Factor-23 (FGF23) and its Fragments in Humans. J Bone Miner Res 2022; 37:1170-1178. [PMID: 35373859 PMCID: PMC9177785 DOI: 10.1002/jbmr.4553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/08/2022] [Accepted: 03/28/2022] [Indexed: 11/06/2022]
Abstract
Relative abundance of fibroblast growth factor-23 (FGF23) measured by the C-terminal (cFGF23, which measures both intact FGF23 and C-terminal fragments) versus intact (iFGF23, measures only intact hormone) assays varies by kidney function in humans. Differential kidney clearance may explain this finding. We measured cFGF23 and iFGF23 in the aorta and bilateral renal veins of 162 patients with essential hypertension undergoing renal angiography. Using multivariable linear regression, we examined factors associated with aorta to renal vein reduction of FGF23 using both assays. Similar parameters and with addition of urine concentrations of cFGF23 and iFGF23 were measured in six Wistar rats. Mean ± standard deviation (SD) age was 54 ± 12 years, 54% were women, and mean creatinine clearance was 72 ± 48 mL/min/100 g. The human kidney reduced the concentrations of both cFGF23 (16% ± 12%) and iFGF23 (21% ± 16%), but reduction was higher for iFGF23. Greater kidney creatinine and PTH reductions were each independently associated with greater reductions of both cFGF23 and iFGF23. The greater kidney reduction of iFGF23 compared to cFGF23 appeared stable and consistent across the range of creatinine clearance evaluated. Kidney clearance was similar, and urine concentrations of both assays were low in the rat models, suggesting kidney metabolism of both cFGF23 and iFGF23. Renal reduction of iFGF23 is higher than that of creatinine and cFGF23. Our data suggest that FGF23 is metabolized by the kidney. However, the major cell types involved in metabolization of FGF23 requires future study. Kidney clearance of FGF23 does not explain differences in C-terminal and intact moieties across the range of kidney function. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Shilpa Sharma
- Division of Nephrology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.,Nephrology Section, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Ronit Katz
- University of Washington, Seattle, WA, USA
| | - Charles Ginsberg
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Alexander Bullen
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, San Diego, CA, USA.,Nephrology Section, Veterans Affairs San Diego Healthcare System, La Jolla, CA, USA
| | - Volker Vallon
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, San Diego, CA, USA.,Nephrology Section, Veterans Affairs San Diego Healthcare System, La Jolla, CA, USA.,Department of Pharmacology, University of California-San Diego, La Jolla, CA, USA
| | - Scott Thomson
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, San Diego, CA, USA.,Nephrology Section, Veterans Affairs San Diego Healthcare System, La Jolla, CA, USA.,Department of Pharmacology, University of California-San Diego, La Jolla, CA, USA
| | - Orson W Moe
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Departments of Internal Medicine and Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Peter W de Leeuw
- Department of Internal Medicine and CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Abraham A Kroon
- Department of Internal Medicine and CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Alfons J H M Houben
- Department of Internal Medicine and CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Joachim H Ix
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, San Diego, CA, USA.,Nephrology Section, Veterans Affairs San Diego Healthcare System, La Jolla, CA, USA
| |
Collapse
|
24
|
Ferreira AC, Mendes M, Silva C, Cotovio P, Aires I, Navarro D, Caeiro F, Ramos R, Salvador R, Correia B, Cabral G, Nolasco F, Ferreira A. Improvement of Mineral and Bone Disorders After Renal Transplantation. Transplantation 2022; 106:e251-e261. [PMID: 35266925 PMCID: PMC9038238 DOI: 10.1097/tp.0000000000004099] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/07/2022] [Accepted: 01/19/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Posttransplant mineral and bone diseases are causes of fractures, and their association with cardiovascular events is being studied. METHODS We analyzed the evolution of biochemical, histological, and imaging parameters pre- and 1 y post-renal transplantation in 69 patients and correlated mineral and bone findings with coronary calcifications. At inclusion and after 12 mo, clinical data and echocardiographic findings were recorded, and laboratory evaluations, radiography of the pelvis and hands, and bone biopsy were performed. Noncontrast cardiac computed tomography was performed during the second evaluation. RESULTS Serum levels of fibroblast growth factor 23 and sclerostin decreased in all patients, parathyroid hormone levels decreased in 89.8% of patients, bone alkaline phosphatase levels decreased in 68.1% of patients, and alpha-Klotho levels increased in 65.2% of patients. More than half of the patients presented with renal osteodystrophy at both biopsies, but histological findings improved: a significant transition from high to normal or low turnover and no significant differences in volume, mineralization defect, or cortical porosity at the 2 evaluations. Alpha-Klotho, sclerostin, and bone alkaline phosphatase shifts affect bone changes. Neither echocardiographic findings nor vascular calcification scores differed between the 2 points. Both the pretransplant period (dialysis vintage, sclerostin, and low bone volume at baseline) and the maintenance of abnormalities in the posttransplant period (high turnover posttransplant) were the most reliable predictors of the severity of the coronary calcification percentile. CONCLUSIONS Renal transplantation improved bone and mineral abnormalities. The pretransplant period determines the severity of calcification.
Collapse
Affiliation(s)
- Ana Carina Ferreira
- Nephrology Department, Hospital Curry Cabral, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- Nova Medical School, Lisbon, Portugal
| | - Marco Mendes
- Nephrology Department, Hospital Curry Cabral, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Cecília Silva
- Nephrology Department, Hospital Curry Cabral, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Patrícia Cotovio
- Nephrology Department, Hospital Curry Cabral, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Inês Aires
- Nephrology Department, Hospital Curry Cabral, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- Nova Medical School, Lisbon, Portugal
| | - David Navarro
- Nephrology Department, Hospital Curry Cabral, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Fernando Caeiro
- Nephrology Department, Hospital Curry Cabral, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Rúben Ramos
- Cardiology Department, Hospital de Santa Marta, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
| | - Rute Salvador
- CEDOC, Tissue Repair and Inflammation Lab, Lisbon, Portugal
| | - Bruna Correia
- CEDOC, Tissue Repair and Inflammation Lab, Lisbon, Portugal
| | | | - Fernando Nolasco
- Nephrology Department, Hospital Curry Cabral, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- Nova Medical School, Lisbon, Portugal
| | - Aníbal Ferreira
- Nephrology Department, Hospital Curry Cabral, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- Nova Medical School, Lisbon, Portugal
| |
Collapse
|
25
|
Quiroga B, Ortiz A, Navarro-González JF, Santamaría R, de Sequera P, Díez J. From cardiorenal syndromes to cardionephrology: a reflection by nephrologists on renocardiac syndromes. Clin Kidney J 2022; 16:19-29. [PMID: 36726435 PMCID: PMC9871856 DOI: 10.1093/ckj/sfac113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Indexed: 02/04/2023] Open
Abstract
Cardiorenal syndromes (CRS) are broadly defined as disorders of the heart and kidneys whereby acute or chronic dysfunction in one organ may induce acute or chronic dysfunction of the other. CRS are currently classified into five categories, mostly based on disease-initiating events and their acuity or chronicity. CRS types 3 and 4 (also called renocardiac syndromes) refer to acute and chronic kidney dysfunction resulting in acute and chronic heart dysfunction, respectively. The notion of renocardiac syndromes has broadened interest in kidney-heart interactions but uncertainty remains in the nephrological community's understanding of the clinical diversity, pathophysiological mechanisms and optimal management approaches of these syndromes. This triple challenge that renocardiac syndromes (and likely other cardiorenal syndromes) pose to the nephrologist can only be faced through a specific and demanding training plan to enhance his/her cardiological scientific knowledge and through an appropriate clinical environment to develop his/her cardiological clinical skills. The first must be the objective of the subspecialty of cardionephrology (or nephrocardiology) and the second must be the result of collaboration with cardiologists (and other specialists) in cardiorenal care units. This review will first consider various aspects of the challenges that renocardiac syndromes pose to nephrologists and, then, will discuss those aspects of cardionephrology and cardiorenal units that can facilitate an effective response to the challenges.
Collapse
Affiliation(s)
| | | | - Juan F Navarro-González
- RICORS2040, Carlos III Institute of Health, Madrid, Spain,Division of Nephrology and Research Unit, University Hospital Nuestra Señora de Candelaria, and University Institute of Biomedical Technologies, University of La Laguna, Santa Cruz de Tenerife, Spain
| | - Rafael Santamaría
- RICORS2040, Carlos III Institute of Health, Madrid, Spain,Division of Nephrology, University Hospital Reina Sofia, Cordoba, Spain,Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Patricia de Sequera
- Department of Nephrology, University Hospital Infanta Leonor, University Complutense of Madrid, Madrid, Spain
| | | |
Collapse
|
26
|
References Values of Soluble α-Klotho Serum Levels Using an Enzyme-Linked Immunosorbent Assay in Healthy Adults Aged 18-85 Years. J Clin Med 2022; 11:jcm11092415. [PMID: 35566540 PMCID: PMC9101232 DOI: 10.3390/jcm11092415] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/21/2022] [Accepted: 04/23/2022] [Indexed: 02/01/2023] Open
Abstract
α-Klotho protein is a powerful predictor of the aging process and lifespan. Although lowered circulating soluble α-Klotho levels have been observed in aged non-healthy individuals, no specific reference values across a wide range of ages and sex using an enzyme-linked immunosorbent assay (ELISA) are available for larger cohorts of healthy individuals. The present analytical cross-sectional study was aimed to establish the reference values of soluble α-Klotho serum levels in healthy adults by age and sex groups. A total of 346 (59% women) healthy individuals aged from 18 to 85 years were recruited. Subjects were divided by sex and age as: (i) young (18−34.9 years), (ii) middle-aged (35−54.9 years), and (iii) senior (55−85 years) individuals. The soluble α-Klotho levels were measured in serum using ELISA. Senior adults were the age-group that presented the lowest soluble α-Klotho serum levels (p < 0.01), with age showing a negative association with soluble α-Klotho serum levels (p < 0.001). No differences between sexes were observed. Therefore, soluble α-Klotho levels were especially decreased—regardless of sex—in our cohort of healthy individuals because of the physiological decline derived from the aging process. We recommend routine assessments of soluble α-Klotho levels using ELISA as a simple and cheap detectable marker of aging that improves quality of life in the elderly.
Collapse
|
27
|
Liu Q, Li S, Yu L, Yin X, Liu X, Ye J, Lu G. CCL5 Suppresses Klotho Expression via p-STAT3/DNA Methyltransferase1-Mediated Promoter Hypermethylation. Front Physiol 2022; 13:856088. [PMID: 35299661 PMCID: PMC8922032 DOI: 10.3389/fphys.2022.856088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/09/2022] [Indexed: 01/19/2023] Open
Abstract
Background Enhanced inflammation and reduced Klotho are common features in chronic kidney disease (CKD). Inflammation induces DNA hypermethylation. This study assessed the performance of inflammatory marker C-C motif chemokine 5 (CCL5) in epigenetic regulation of Klotho expression. Methods Fifty CKD patients and 25 matched controls were enrolled, and serum CCL5 level, sKlotho level, and DNA methylation were evaluated in these subjects. A renal interstitial fibrosis (RIF) model with CKD was induced in mice via unilateral ureteral obstruction (UUO) in vivo and human proximal tubular epithelial (HK-2) cells treated with CCL5 in vitro. 5-aza-2′-deoxycytidine (5-Aza), a DNA methyltransferase inhibitor was given to UUO mice. Hematoxylin and eosin (HE) and Masson trichrome staining were adopted to evaluate renal pathological changes. Methylation-specific PCR was performed to assess DNA methylation of Klotho promoter in the peripheral blood leucocytes (PBLs) from CKD patients and obstructive kidney from UUO mice. CCL5, Klotho, and DNA methyltransferases (DNMTs) were determined by ELISAs, immunofluorescence, or western blotting. HK-2 cells were exposed to CCL5 with or without 5-Aza and stattic, a p-signal transducer and activator of transcription 3 (STAT3) inhibitor, and expressions of p-STAT3, DNMT1, and Klotho were determined by western blotting. Results CCL5 upregulation concomitant with Klotho downregulation in serum and global DNA methylation in PBLs were observed in CKD samples. UUO contributed to severe renal interstitial fibrosis and enhanced expressions of fibrotic markers. Moreover, UUO increased the CCL5 level, induced Klotho promoter methylation, suppressed Klotho level, activated p-STAT3 signaling, and upregulated DNMT1 level. A similar observation was made in HK-2 cells treated with CCL5. More importantly, 5-Aza inhibited UUO-induced Klotho hypermethylation, reversed Klotho, downregulated p-STAT3 expressions, and ameliorated RIF in vivo. The consistent findings in vitro were also obtained in HK-2 cells exposed to 5-Aza and stattic. Conclusion The CCL5/p-STAT3/DNMT1 axis is implicated in epigenetic regulation of Klotho expression in CKD. This study provides novel therapeutic possibilities for reversal of Klotho suppression by CKD.
Collapse
Affiliation(s)
- QiFeng Liu
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - ShaSha Li
- Clinical Research & Lab Centre, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - LiXia Yu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - XiaoYa Yin
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Xi Liu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - JianMing Ye
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - GuoYuan Lu
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
28
|
Drew DA, Katz R, Kritchevsky S, Ix JH, Shlipak MG, Newman AB, Hoofnagle AN, Fried LF, Sarnak M, Gutiérrez OM, Semba RD, Neyra JA. Soluble Klotho and Incident Hypertension. Clin J Am Soc Nephrol 2021; 16:1502-1511. [PMID: 34556498 PMCID: PMC8498995 DOI: 10.2215/cjn.05020421] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/22/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND OBJECTIVES Hypertension is associated with significant morbidity and mortality despite effective antihypertensive therapies. Soluble klotho is a circulating protein that in preclinical studies is protective against the development of hypertension. There are limited studies of klotho and blood pressure in humans. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Within the Health, Aging, and Body Composition Study, a cohort of well-functioning older adults, soluble klotho was measured in serum. We evaluated the cross-sectional and longitudinal association between klotho and blood pressure, prevalent hypertension, incident hypertension, and BP trajectories. Analyses were adjusted for demographics, cardiovascular disease and kidney disease risk factors, and measures of mineral metabolism including calcium, phosphate, parathyroid hormone, 25(OH) vitamin D, and fibroblast growth factor 23. RESULTS The median klotho concentration was 630 pg/ml (478-816, 25th to 75th percentile). Within the cohort, 2093 (76%) of 2774 participants had prevalent hypertension and 476 (70%) of the remaining 681 developed incident hypertension. There was no association between klotho and prevalent hypertension or baseline systolic BP, but higher klotho was associated with higher baseline diastolic BP (fully adjusted β=0.92 mmHg, 95% confidence interval, 0.24 to 1.60 mmHg, higher per two-fold higher klotho). Higher baseline serum klotho levels were significantly associated with a lower rate of incident hypertension (fully adjusted hazard ratio, 0.80; 95% confidence interval, 0.69 to 0.93 for every two-fold higher klotho). Higher klotho was also associated with lower subsequent systolic BP and diastolic BP (-0.16, 95% confidence interval, -0.31 to -0.01, mmHg lower systolic BP per year and -0.10, 95% confidence interval, -0.18 to -0.02, mmHg lower diastolic BP per year, for each two-fold higher klotho). CONCLUSIONS Higher klotho is associated with higher baseline diastolic but not systolic BP, a lower risk of incident hypertension, and lower BP trajectories during follow-up.
Collapse
Affiliation(s)
- David A. Drew
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Ronit Katz
- Kidney Research Institute, University of Washington, Seattle, Washington
| | - Stephen Kritchevsky
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Joachim H. Ix
- Division of Nephrology-Hypertension, University of California San Diego School of Medicine, San Diego, California
| | - Michael G. Shlipak
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Health Care System, San Francisco, California,Kidney Health Research Collaborative, University of California San Francisco, San Francisco, California
| | - Anne B. Newman
- Kidney Health Research Collaborative, University of California San Francisco, San Francisco, California
| | | | - Linda F. Fried
- University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania,Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Mark Sarnak
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Orlando M. Gutiérrez
- Medicine - Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Richard D. Semba
- Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Javier A. Neyra
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas,The Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas,Division of Nephrology, University of Kentucky Medical Center, Lexington, Kentucky
| |
Collapse
|
29
|
Freundlich M, Gamba G, Rodriguez-Iturbe B. Fibroblast growth factor 23-Klotho and hypertension: experimental and clinical mechanisms. Pediatr Nephrol 2021; 36:3007-3022. [PMID: 33230698 PMCID: PMC7682775 DOI: 10.1007/s00467-020-04843-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 10/07/2020] [Accepted: 10/23/2020] [Indexed: 12/11/2022]
Abstract
Hypertension (HTN) and chronic kidney disease (CKD) are increasingly recognized in pediatric patients and represent risk factors for cardiovascular morbidity and mortality later in life. In CKD, enhanced tubular sodium reabsorption is a leading cause of HTN due to augmented extracellular fluid volume expansion. The renin-angiotensin-aldosterone system (RAAS) upregulates various tubular sodium cotransporters that are also targets of the hormone fibroblast growth factor 23 (FGF23) and its co-receptor Klotho. FGF23 inhibits the activation of 1,25-dihydroxyvitamin D that is a potent suppressor of renin biosynthesis. Here we review the complex interactions and disturbances of the FGF23-Klotho axis, vitamin D, and the RAAS relevant to blood pressure regulation and discuss the therapeutic strategies aimed at mitigating their pathophysiologic contributions to HTN.
Collapse
Affiliation(s)
- Michael Freundlich
- Department of Pediatrics, Division of Pediatric Nephrology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Gerardo Gamba
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Department of Nephrology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Bernardo Rodriguez-Iturbe
- Department of Nephrology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Department of Nephrology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| |
Collapse
|
30
|
Gergei I, Zheng J, Andlauer TFM, Brandenburg V, Mirza-Schreiber N, Müller-Myhsok B, Krämer BK, Richard D, Falk L, Movérare-Skrtic S, Ohlsson C, Smith GD, März W, Voelkl J, Tobias JH. GWAS META-analysis followed by MENDELIAN randomisation revealed potential control mechanisms for circulating α-klotho levels. Hum Mol Genet 2021; 31:792-802. [PMID: 34542150 PMCID: PMC8895756 DOI: 10.1093/hmg/ddab263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/08/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The protein α-Klotho acts as transmembrane the co-receptor for fibroblast growth factor 23 (FGF-23) and is a key regulator of phosphate homeostasis. However, α-Klotho also exists in a circulating form, with pleiotropic, but incompletely understood functions and regulation. Therefore, we undertook a GWAS meta-analysis followed by Mendelian randomisation (MR) of circulating α-Klotho levels. METHODS Plasma α-Klotho levels were measured by ELISA in the LURIC and ALSPAC (mothers) cohorts, followed by a GWAS meta-analysis in 4376 individuals across the two cohorts. RESULTS Six signals at five loci were associated with circulating α-Klotho levels at genome-wide significance (p < 5 × 10-8), namely ABO, KL, FGFR1, and two post-translational modification genes, B4GALNT3 and CHST9. Together, these loci explained > 9% of the variation in circulating α-Klotho levels. MR analyses revealed no causal relationships between α-Klotho and renal function, FGF-23-dependent factors such as vitamin D and phosphate levels, or bone mineral density. The screening for genetic correlations with other phenotypes, followed by targeted MR suggested causal effects of liability of Crohn's disease risk [IVW beta = 0.059 (95% CI 0.026, 0.093)] and low-density lipoprotein cholesterol (LDL-C) levels [-0.198, (-0.332, -0.063)] on α-Klotho. CONCLUSIONS Our GWAS findings suggest that two enzymes involved in post-translational modification, B4GALNT3 and CHST9, contribute to genetic influences on α-Klotho levels, presumably by affecting protein turnover and stability. Subsequent evidence from MR analyses on α-Klotho levels suggest regulation by mechanisms besides phosphate-homeostasis and raise the possibility of cross-talk with FGF19- and FGF21-dependent pathways, respectively.
Collapse
Affiliation(s)
- Ingrid Gergei
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), University Medical Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Therapeutic Area Cardiovascular Medicine, Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Jie Zheng
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, United Kingdom.,Population Health Science, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, United Kingdom
| | - Till F M Andlauer
- Max Planck Institute of Psychiatry, Munich, Germany.,Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | | | | | - Bertram Müller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Bernhard K Krämer
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), University Medical Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,European Center for Angioscience ECAS, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Daniel Richard
- Department of Human Evolutionary Biology, Harvard University, USA
| | - Louise Falk
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, United Kingdom
| | - Sofia Movérare-Skrtic
- University of Gothenburg, Sahlgrenska Osteoporosis Centre, CBAR, Institute of Medicine, Department of Internal Medicine and Clinical Nutrition, Gothenburg, Sweden
| | - Claes Ohlsson
- University of Gothenburg, Sahlgrenska Osteoporosis Centre, CBAR, Institute of Medicine, Department of Internal Medicine and Clinical Nutrition, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital, Department of Drug Treatment, Gothenburg, Sweden
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, United Kingdom.,Population Health Science, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, United Kingdom
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), University Medical Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Mannheim, Germany.,Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria
| | - Jakob Voelkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria.,Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Jonathan H Tobias
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, United Kingdom.,Musculoskeletal Research Unit, Translational HeaalthLevel 1 Learning and Research Building, Southmead Hospital, Bristol, United Kingdom
| |
Collapse
|
31
|
Epstein M, Freundlich M. The intersection of Mineralocorticoid Receptor (MR) activation and the FGF23 - Klotho cascade. A Duopoly that promotes renal and cardiovascular injury. Nephrol Dial Transplant 2021; 37:211-221. [PMID: 34459924 DOI: 10.1093/ndt/gfab254] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Indexed: 12/17/2022] Open
Abstract
The nexus of CKD and cardiovascular disease (CVD) amplifies the morbidity and mortality of CKD, emphasizing the need for defining and establishing therapeutic initiatives to modify and abrogate the progression of CKD and concomitant CV risks. In addition to the traditional CV risk factors, disturbances of mineral metabolism are specific risk factors that contribute to the excessive CV mortality in patients with CKD. These risk factors include dysregulations of circulating factors that modulate phosphate metabolism including fibroblast growth factor 23 (FGF23) and soluble Klotho. Reduced circulating levels and suppressed renal klotho expression may be associated with adverse outcomes in CKD patients. While elevated circulating concentrations or locally produced FGF23 in the strained heart exert pro-hypertrophic mechanisms on the myocardium, Klotho attenuates tissue fibrosis, progression of CKD, cardiomyopathy, endothelial dysfunction, vascular stiffness, and vascular calcification. Mineralocorticoid receptor (MR) activation in non-classical targets, mediated by aldosterone and other ligands, amplifies CVD in CKD. In concert, we detail how the interplay of elevated FGF23, activation of the MR, and concomitant reductions of circulating Klotho in CKD, may potentiate each other's deleterious effects on kidney and the heart, thereby contributing to the initiation and progression of kidney and cardiac functional deterioration, acting through multipronged albeit complementary mechanistic pathways.
Collapse
Affiliation(s)
- Murray Epstein
- Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michael Freundlich
- Division of Pediatric Nephrology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
32
|
Liu Q, Yu L, Yin X, Ye J, Li S. Correlation Between Soluble Klotho and Vascular Calcification in Chronic Kidney Disease: A Meta-Analysis and Systematic Review. Front Physiol 2021; 12:711904. [PMID: 34483963 PMCID: PMC8414804 DOI: 10.3389/fphys.2021.711904] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/14/2021] [Indexed: 11/29/2022] Open
Abstract
Background: The correlation between soluble Klotho (sKlotho) level and vascular calcification (VC) in patients with chronic kidney disease (CKD) remains controversial. Using meta-analysis, we aimed to address this controversy and assess the feasibility of applying sKlotho as a biomarker for VC. Methods: Medical electronic databases were thoroughly searched for eligible publications on the association between sKlotho level and VC in CKD patients. Effectors, including correlation coefficients (r), odds ratios (ORs), hazard ratio (HR) or β-values, and 95% confidence intervals (CIs) were extracted and combined according to study design or effector calculation method. Pooled effectors were generated using both random-effects models and fixed-effects models according to I 2-value. Origin of heterogeneity was explored by sensitivity analysis and subgroup analysis. Results: Ten studies with 1,204 participants from a total of 1,199 publications were eligible and included in this meta-analysis. The combined correlation coefficient (r) was [-0.33 (-0.62, -0.04)] with significant heterogeneity (I 2 = 89%, p < 0.001) based on Spearman correlation analysis, and this significant association was also demonstrated in subgroups. There was no evidence of publication bias. The combined OR was [3.27 (1.70, 6.30)] with no evidence of heterogeneity (I 2 = 0%, p = 0.48) when sKlotho was treated as a categorical variable or [1.05 (1.01, 1.09)] with moderate heterogeneity (I 2 = 63%, p = 0.10) when sKlotho was treated as a continuous variable based on multivariate logistic regression. No significant association was observed and the pooled OR was [0.29 (0.01, 11.15)] with high heterogeneity (I 2 = 96%, p < 0.001) according to multivariate linear regression analysis. There was an inverse association between sKlotho and parathyroid hormone levels. The combined coefficient (r) was [-0.20 (-0.40, -0.01)] with significant heterogeneity (I 2 = 86%, p < 0.001), and without obvious publication bias. No significant association was found between sKlotho and calcium or phosphate levels. Conclusion: There exists a significant association between decreased sKlotho level and increased risk of VC in CKD patients. This raises the possibility of applying sKlotho as a biomarker for VC in CKD populations. Large, prospective, well-designed studies or interventional clinical trials are required to validate our findings.
Collapse
Affiliation(s)
- QiFeng Liu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - LiXia Yu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - XiaoYa Yin
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - JianMing Ye
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - ShaSha Li
- Clinical Research & Lab Centre, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| |
Collapse
|
33
|
Kanbay M, Demiray A, Afsar B, Covic A, Tapoi L, Ureche C, Ortiz A. Role of Klotho in the Development of Essential Hypertension. Hypertension 2021; 77:740-750. [PMID: 33423524 DOI: 10.1161/hypertensionaha.120.16635] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Klotho has antiaging properties, and serum levels decrease with physiological aging and aging-related diseases, such as hypertension, cardiovascular, and chronic kidney disease. Klotho deficiency in mice results in accelerated aging and cardiovascular injury, whereas Klotho supplementation slows down the progression of aging-related diseases. The pleiotropic functions of Klotho include, but are not limited to, inhibition of insulin/IGF-1 (insulin-like growth factor 1) and WNT (wingless-related integration site) signaling pathways, suppression of oxidative stress and aldosterone secretion, regulation of calcium-phosphate homeostasis, and modulation of autophagy with inhibition of apoptosis, fibrosis, and cell senescence. Accumulating evidence shows an interconnection between Klotho deficiency and hypertension, and Klotho gene polymorphisms are associated with hypertension in humans. In this review, we critically review the current understanding of the role of Klotho in the development of essential hypertension and the most important underlying pathways involved, such as the FGF23 (fibroblast growth factor 23)/Klotho axis, aldosterone, Wnt5a/RhoA, and SIRT1 (Sirtuin1). Based on this critical review, we suggest avenues for further research.
Collapse
Affiliation(s)
- Mehmet Kanbay
- From the Division of Nephrology, Department of Medicine (M.K.), Koc University School of Medicine, Istanbul, Turkey
| | - Atalay Demiray
- Department of Medicine (A.D.), Koc University School of Medicine, Istanbul, Turkey
| | - Baris Afsar
- Division of Nephrology, Department of Internal Medicine, Suleyman Demirel University School of Medicine, Isparta Turkey (B.A.)
| | - Adrian Covic
- Department of Nephrology, Grigore T. Popa University of Medicine, Iasi, Romania (A.C., L.T., C.U.)
| | - Laura Tapoi
- Department of Nephrology, Grigore T. Popa University of Medicine, Iasi, Romania (A.C., L.T., C.U.)
| | - Carina Ureche
- Department of Nephrology, Grigore T. Popa University of Medicine, Iasi, Romania (A.C., L.T., C.U.)
| | - Alberto Ortiz
- Cardiovascular Diseases Institute, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.O.)
- IIS-Fundacion Jimenez Diaz, Department of Medicine, School of Medicine, Universidad Autonoma de Madrid, Spain (A.O.)
| |
Collapse
|
34
|
Hu PP, Bao JF, Li A. Roles for fibroblast growth factor-23 and α-Klotho in acute kidney injury. Metabolism 2021; 116:154435. [PMID: 33220250 DOI: 10.1016/j.metabol.2020.154435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/08/2020] [Accepted: 11/13/2020] [Indexed: 12/21/2022]
Abstract
Acute kidney injury is a global disease with high morbidity and mortality. Recent studies have revealed that the fibroblast growth factor-23-α-Klotho axis is closely related to chronic kidney disease, and has multiple biological functions beyond bone-mineral metabolism. However, although dysregulation of fibroblast growth factor-23-α-Klotho has been observed in acute kidney injury, the role of fibroblast growth factor-23-α-Klotho in the pathophysiology of acute kidney injury remains largely unknown. In this review, we describe recent findings regarding fibroblast growth factor-23-α-Klotho, which is mainly involved in inflammation, oxidative stress, and hemodynamic disorders. Further, based on these recent results, we put forth novel insights regarding the relationship between the fibroblast growth factor-23-α-Klotho axis and acute kidney injury, which may provide new therapeutic targets for treating acute kidney injury.
Collapse
Affiliation(s)
- Pan-Pan Hu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China
| | - Jing-Fu Bao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China
| | - Aiqing Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China; Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China.
| |
Collapse
|
35
|
Kakar RS, Pastor JV, Moe OW, Ambrosio F, Castaldi D, Sanders LH. Peripheral Klotho and Parkinson's Disease. Mov Disord 2021; 36:1274-1276. [PMID: 33629770 DOI: 10.1002/mds.28530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/25/2021] [Indexed: 11/06/2022] Open
Affiliation(s)
- Rumit S Kakar
- Old Dominion University Norfolk, Norfolk, Virginia, USA
| | - Johanne V Pastor
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Orson W Moe
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | | | - Laurie H Sanders
- Department of Neurology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
36
|
Jadhav S, Tripathi S, Chandrekar A, Waikar SS, Hsiao LL. A novel antibody for the detection of alternatively spliced secreted KLOTHO isoform in human plasma. PLoS One 2021; 16:e0245614. [PMID: 33481901 PMCID: PMC7822350 DOI: 10.1371/journal.pone.0245614] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 01/04/2021] [Indexed: 11/19/2022] Open
Abstract
αKlotho is primarily known to express as a transmembrane protein. Proteolytic cleavage results in shedding of the extracellular domain which enters systemic circulation. A truncated form of αKlotho resulting from alternative splicing of the αKLOTHO transcript exists and is believed to be secreted, thereby also entering systemic circulation. Existing ELISA methods fail to distinguish between the two circulating isoforms resulting in inconsistencies in assessing circulating αKlotho levels. We have exploited a unique 15aa peptide sequence present in the alternatively spliced secreted isoform to generate an antibody and show that it is able to specifically detect only the secreted Klotho isoform in human plasma. This finding will facilitate in distinguishing the levels of different circulating Klotho isoforms in health and disease and enhance their potential to serve as a biomarker for CKD and other conditions.
Collapse
Affiliation(s)
- Shreyas Jadhav
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sudipta Tripathi
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Anil Chandrekar
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sushrut S. Waikar
- Section of Nephrology, Boston University Medical Center, Boston, Massachusetts, United States of America
| | - Li-Li Hsiao
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
37
|
Neyra JA, Hu MC, Moe OW. Klotho in Clinical Nephrology: Diagnostic and Therapeutic Implications. Clin J Am Soc Nephrol 2020; 16:162-176. [PMID: 32699047 PMCID: PMC7792642 DOI: 10.2215/cjn.02840320] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
αKlotho (called Klotho here) is a membrane protein that serves as the coreceptor for the circulating hormone fibroblast growth factor 23 (FGF23). Klotho is also cleaved and released as a circulating substance originating primarily from the kidney and exerts a myriad of housekeeping functions in just about every organ. The vital role of Klotho is shown by the multiorgan failure with genetic deletion in rodents, with certain features reminiscent of human disease. The most common causes of systemic Klotho deficiency are AKI and CKD. Preclinical data on Klotho biology have advanced considerably and demonstrated its potential diagnostic and therapeutic value; however, multiple knowledge gaps exist in the regulation of Klotho expression, release, and metabolism; its target organs; and mechanisms of action. In the translational and clinical fronts, progress has been more modest. Nonetheless, Klotho has potential clinical applications in the diagnosis of AKI and CKD, in prognosis of progression and extrarenal complications, and finally, as replacement therapy for systemic Klotho deficiency. The overall effect of Klotho in clinical nephrology requires further technical advances and additional large prospective human studies.
Collapse
Affiliation(s)
- Javier A. Neyra
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Division of Nephrology, Bone and Mineral Metabolism, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky
| | - Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Orson W. Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
38
|
Prikryl P, Satrapova V, Frydlova J, Hruskova Z, Zima T, Tesar V, Vokurka M. Mass spectrometry-based proteomic exploration of the small urinary extracellular vesicles in ANCA-associated vasculitis in comparison with total urine. J Proteomics 2020; 233:104067. [PMID: 33307252 DOI: 10.1016/j.jprot.2020.104067] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/11/2020] [Accepted: 11/29/2020] [Indexed: 01/07/2023]
Abstract
ANCA-associated vasculitis (AAV) is a rare, but potentially severe autoimmune disease, even nowadays displaying increased mortality and morbidity. Finding early biomarkers of activity and prognosis is thus very important. Small extracellular vesicles (EVs) isolated from urine can be considered as a non-invasive source of biomarkers. We evaluated several protocols for urinary EV isolation. To eliminate contaminating non-vesicular proteins due to AAV associated proteinuria we used proteinase K treatment. We investigated the differences in proteomes of small EVs of patients with AAV compared to healthy controls by label-free LC-MS/MS. In parallel, we performed an analogous proteomic analysis of urine samples from identical patients. The study results showed significant differences and similarities in both EV and urine proteome, the latter one being highly affected by proteinuria. Using bioinformatics tools we explored differentially changed proteins and their related pathways with a focus on the pathophysiology of AAV. Our findings indicate significant regulation of Golgi enzymes, such as MAN1A1, which can be involved in T cell activation by N-glycans glycosylation and may thus play a key role in pathogenesis and diagnosis of AAV. SIGNIFICANCE: The present study explores for the first time the changes in proteomes of small extracellular vesicles and urine of patients with renal ANCA-associated vasculitis compared to healthy controls by label-free LC-MS/MS. Isolation of vesicles from proteinuric urine samples has been modified to minimize contamination by plasma proteins and to reduce co-isolation of extraluminal proteins. Differentially changed proteins and their related pathways with a role in the pathophysiology of AAV were described and discussed. The results could be helpful for the research of potential biomarkers in renal vasculitis associated with ANCA.
Collapse
Affiliation(s)
- Petr Prikryl
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Veronika Satrapova
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jana Frydlova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Zdenka Hruskova
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Tomas Zima
- Institute of Clinical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Vladimir Tesar
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Martin Vokurka
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
39
|
Ortiz A. The unaccomplished mission of reducing mortality in patients on kidney replacement therapy. Clin Kidney J 2020; 13:948-951. [PMID: 33391738 PMCID: PMC7769530 DOI: 10.1093/ckj/sfaa235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 10/29/2020] [Indexed: 12/21/2022] Open
Abstract
Six years ago, a comprehensive review by the EURECA-m working group of the ERA-EDTA thoroughly addressed the drivers of mortality in patients with end-stage kidney disease. Not unexpectedly, the key global driver of early death in these patients was the lack of access to kidney replacement therapy. However, and contrary to the expectations of non-nephrologists, mortality was still high when kidney replacement therapy was provided. This was due to excess cardiovascular and non-cardiovascular mortality, and the need to further characterize correctable risk factors and eventually test the impact of correcting them was emphasized. In this issue of ckj, seven reports address risk factors for death in non-dialysis chronic kidney disease (CKD), dialysis and kidney transplant patients. They characterize irreversible (e.g. sex; age; genetic variants of the KL gene encoding the anti-ageing protein Klotho) and reversible (obesity; mineral and bone disorder parameters; anti-depressant drugs, especially those that increase the QT; amputation; public health investments) factors associated with mortality of CKD patients on or off kidney replacement therapy.
Collapse
Affiliation(s)
- Alberto Ortiz
- IIS-Fundacion Jimenez Diaz UAM and School of Medicine, UAM, Madrid, Spain
- GEENDIAB, Madrid, Spain
- REDINREN, Madrid, Spain
| |
Collapse
|
40
|
Yi HJ, Lee JB, Lee KP, Oh YI, Song KH, Seo KW. Investigation on urinary and serum alpha klotho in dogs with chronic kidney disease. BMC Vet Res 2020; 16:246. [PMID: 32677951 PMCID: PMC7364505 DOI: 10.1186/s12917-020-02458-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/06/2020] [Indexed: 01/16/2023] Open
Abstract
Background As a co-receptor for fibroblast growth factor 23, klotho plays a pivotal role in phosphate metabolism. The kidney is known to be the main source of soluble alpha-klotho and the principal regulator of its concentration. Previous studies in human participants showed that the concentration of soluble alpha-klotho in serum and urine decreased in chronic kidney disease (CKD) patients. However, no previous study has assessed soluble alpha-klotho levels in dogs. This study aimed to measure serum and urinary alpha-klotho levels in CKD dogs and identify their associations with International Renal Interest Society (IRIS) CKD stages and other parameters known to be associated with CKD. Results Serum and urinary alpha klotho concentrations were measured by a commercially available canine-specific sandwich enzyme-linked immunosorbent assay kit and compared between groups by a nonparametric Kruskal–Wallis test. Spearman’s correlation coefficient was used to evaluate the relationships between variables. A stepwise multiple regression analysis was performed to estimate the effects of independent predictors on klotho concentrations. The urine klotho-to-creatinine ratio (UrKl/Cr) was significantly lower in stage 3 dogs than the control group and was significantly lower in dogs with stage 3 and 4 CKD than in those with stage 1 and 2 disease. UrKl/Cr was negatively correlated with serum symmetric dimethylarginine (sSDMA), blood urea nitrogen (BUN), creatinine, and phosphorus concentration. Serum alpha-klotho concentration in dogs with stages 2 and 3 CKD was significantly lower than those in the control group. There was no significant correlation between serum alpha-klotho and BUN, creatinine, and phosphorus concentrations. No statistically significant differences were observed in UrKl/Cr and serum alpha-klotho concentration between groups based on sex, age, urine protein-to-creatinine ratio (UPC), or blood pressure. Conclusions UrKl/Cr decreased in dogs with advanced CKD, and it was negatively correlated with sSDMA, BUN, creatinine, and phosphorus concentrations. Thus, klotho is associated with CKD and its clinical consequences, including CKD-mineral bone disorder, in dogs. Although serum klotho concentration was negatively correlated with sSDMA levels, it was not apparently related to IRIS CKD stage or other parameters known to be associated with CKD.
Collapse
Affiliation(s)
- Hong Jae Yi
- VMTH of College of Veterinary Medicine, Chungnam National University, N13-2, #308, 99 Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Jong Bok Lee
- College of Veterinary Medicine, Kangwon National University, Chuncheon, Korea
| | - Kyu Pil Lee
- VMTH of College of Veterinary Medicine, Chungnam National University, N13-2, #308, 99 Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Ye-In Oh
- VMTH of College of Veterinary Medicine, Chungnam National University, N13-2, #308, 99 Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Kun Ho Song
- VMTH of College of Veterinary Medicine, Chungnam National University, N13-2, #308, 99 Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Kyoung Won Seo
- VMTH of College of Veterinary Medicine, Chungnam National University, N13-2, #308, 99 Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea.
| |
Collapse
|
41
|
Thongprayoon C, Neyra JA, Hansrivijit P, Medaura J, Leeaphorn N, Davis PW, Kaewput W, Bathini T, Salim SA, Chewcharat A, Aeddula NR, Vallabhajosyula S, Mao MA, Cheungpasitporn W. Serum Klotho in Living Kidney Donors and Kidney Transplant Recipients: A Meta-Analysis. J Clin Med 2020; 9:1834. [PMID: 32545510 PMCID: PMC7355868 DOI: 10.3390/jcm9061834] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/07/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023] Open
Abstract
α-Klotho is a known anti-aging protein that exerts diverse physiological effects, including phosphate homeostasis. Klotho expression occurs predominantly in the kidney and is significantly decreased in patients with chronic kidney disease. However, changes in serum klotho levels and impacts of klotho on outcomes among kidney transplant (KTx) recipients and kidney donors remain unclear. A literature search was conducted using MEDLINE, EMBASE, and Cochrane Database from inception through October 2019 to identify studies evaluating serum klotho levels and impacts of klotho on outcomes among KTx recipients and kidney donors. Study results were pooled and analyzed utilizing a random-effects model. Ten cohort studies with a total of 431 KTx recipients and 5 cohort studies with a total of 108 living kidney donors and were identified. After KTx, recipients had a significant increase in serum klotho levels (at 4 to 13 months post-KTx) with a mean difference (MD) of 243.11 pg/mL (three studies; 95% CI 67.41 to 418.81 pg/mL). Although KTx recipients had a lower serum klotho level with a MD of = -234.50 pg/mL (five studies; 95% CI -444.84 to -24.16 pg/mL) compared to healthy unmatched volunteers, one study demonstrated comparable klotho levels between KTx recipients and eGFR-matched controls. Among kidney donors, there was a significant decrease in serum klotho levels post-nephrectomy (day 3 to day 5) with a mean difference (MD) of -232.24 pg/mL (three studies; 95% CI -299.41 to -165.07 pg/mL). At one year following kidney donation, serum klotho levels remained lower than baseline before nephrectomy with a MD of = -110.80 pg/mL (two studies; 95% CI 166.35 to 55.24 pg/mL). Compared to healthy volunteers, living kidney donors had lower serum klotho levels with a MD of = -92.41 pg/mL (two studies; 95% CI -180.53 to -4.29 pg/mL). There is a significant reduction in serum klotho levels after living kidney donation and an increase in serum klotho levels after KTx. Future prospective studies are needed to assess the impact of changes in klotho on clinical outcomes in KTx recipients and living kidney donors.
Collapse
Affiliation(s)
- Charat Thongprayoon
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA;
| | - Javier A. Neyra
- Division of Nephrology, Bone and Mineral Metabolism, Department of Internal Medicine, University of Kentucky, Lexington, KY 40506, USA;
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, TX 75390, USA
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Panupong Hansrivijit
- Department of Internal Medicine, University of Pittsburgh Medical Center Pinnacle, Harrisburg, PA 17105, USA;
| | - Juan Medaura
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.M.); (P.W.D.); (S.A.S.)
| | - Napat Leeaphorn
- Renal Transplant Program, University of Missouri-Kansas City School of Medicine/Saint Luke’s Health System, Kansas City, MO 64110, USA;
| | - Paul W. Davis
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.M.); (P.W.D.); (S.A.S.)
| | - Wisit Kaewput
- Department of Military and Community Medicine, Phramongkutklao College of Medicine, Bangkok 10400, Thailand;
| | - Tarun Bathini
- Department of Internal Medicine, University of Arizona, Tucson, AZ 85721, USA;
| | - Sohail Abdul Salim
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.M.); (P.W.D.); (S.A.S.)
| | - Api Chewcharat
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA;
| | - Narothama Reddy Aeddula
- Division of Nephrology, Department of Medicine, Deaconess Health System, Evansville, IN 47710, USA;
| | | | - Michael A. Mao
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Wisit Cheungpasitporn
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.M.); (P.W.D.); (S.A.S.)
| |
Collapse
|
42
|
Albuminuria Downregulation of the Anti-Aging Factor Klotho: The Missing Link Potentially Explaining the Association of Pathological Albuminuria with Premature Death. Adv Ther 2020; 37:62-72. [PMID: 32236874 DOI: 10.1007/s12325-019-01180-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Indexed: 12/17/2022]
Abstract
Ten percent of the adult population has chronic kidney disease (CKD), which is diagnosed when the glomerular filtration rate (GFR) is below 60 mL/min per 1.73 m2 or when albuminuria is above 30 mg/day. The numerical thresholds were chosen because they are associated with an increased risk of CKD progression or premature death within a wider scenario of accelerated aging. Indeed, CKD is one of the fastest growing causes of death worldwide. A decreased GFR is associated with the accumulation of uraemic toxins that may promote tissue and organ damage. However, CKD may be diagnosed when the GFR is completely normal, as long as there is pathological albuminuria. A key unanswered question to stem the rise of CKD-associated deaths is whether the association between isolated albuminuria (when the GFR is normal) and premature death is causal. The recent demonstration that albuminuria per se directly suppresses the production of the anti-aging factor Klotho by kidney tubular cells may be one of the first steps to address the causality of the albuminuria-premature death-accelerated aging association. This hypothesis should be tested in interventional studies that should draw from translational science advances. Thus, the observation that albuminuria decreases Klotho production through epigenetic mechanisms implies that Klotho downregulation may persist after the correction of albuminuria, and innovative therapeutic approaches are needed to restore Klotho production. On the basis of recent literature, these may include manipulation of NF-kappaB regulators such as B cell lymphoma 3 protein (BCL-3), and epigenetic regulators such as histone deacetylases, or the repurposing of drugs such as pentoxifylline.
Collapse
|
43
|
Sanchez-Niño MD, Fernandez-Fernandez B, Ortiz A. Klotho, the elusive kidney-derived anti-ageing factor. Clin Kidney J 2020; 13:125-127. [PMID: 32297880 PMCID: PMC7147303 DOI: 10.1093/ckj/sfz125] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022] Open
Abstract
Chronic kidney disease (CKD) is one of the fastest growing causes of death worldwide. Only early diagnosis will allow prevention of both CKD progression and the negative impact of CKD on all-cause and cardiovascular mortality. Klotho is a protein produced by the kidneys that has anti-ageing and phosphaturic properties, preventing excess positive phosphate balance. There is evidence that Klotho downregulation is one of the earliest consequences of kidney injury. Thus the development of reliable assays to monitor Klotho levels may allow an early diagnosis of CKD and monitoring the impact of therapies aimed at preserving Klotho expression or at preventing CKD progression. However, the performance of Klotho assays has been suboptimal so far. In this issue of Clinical Kidney Journal, Neyra et al. explore methods to improve the reliability of Klotho assays.
Collapse
Affiliation(s)
- Maria Dolores Sanchez-Niño
- IIS-Fundacion Jimenez Diaz, School of Medicine, Universidad Autonoma de Madrid; Fundacion Renal Iñigo Alvarez de Toledo-IRSIN and REDINREN, Madrid, Spain
| | - Beatriz Fernandez-Fernandez
- IIS-Fundacion Jimenez Diaz, School of Medicine, Universidad Autonoma de Madrid; Fundacion Renal Iñigo Alvarez de Toledo-IRSIN and REDINREN, Madrid, Spain
| | - Alberto Ortiz
- IIS-Fundacion Jimenez Diaz, School of Medicine, Universidad Autonoma de Madrid; Fundacion Renal Iñigo Alvarez de Toledo-IRSIN and REDINREN, Madrid, Spain
| |
Collapse
|
44
|
Lim K, Halim A, Lu TS, Ashworth A, Chong I. Klotho: A Major Shareholder in Vascular Aging Enterprises. Int J Mol Sci 2019; 20:E4637. [PMID: 31546756 PMCID: PMC6770519 DOI: 10.3390/ijms20184637] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/11/2019] [Accepted: 09/13/2019] [Indexed: 01/08/2023] Open
Abstract
Accelerated vascular aging is a condition that occurs as a complication of several highly prevalent inflammatory conditions such as chronic kidney disease, cancer, HIV infection and diabetes. Age-associated vascular alterations underlie a continuum of expression toward clinically overt cardiovascular disease. This has contributed to the striking epidemiologic transition whereby such noncommunicable diseases have taken center stage as modern-day global epidemics and public health problems. The identification of α-Klotho, a remarkable protein that confers powerful anti-aging properties has stimulated significant interest. In fact, emerging data have provided fundamental rationale for Klotho-based therapeutic intervention for vascular diseases and multiple other potential indications. However, the application of such discoveries in Klotho research remains fragmented due to significant gaps in our molecular understanding of Klotho biology, as well as hurdles in clinical research and experimental barriers that must first be overcome. These advances will be critical to establish the scientific platform from which future Klotho-based interventional trials and therapeutic enterprises can be successfully launched.
Collapse
Affiliation(s)
- Kenneth Lim
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- MGH Renal Associates, 165 Cambridge Street, Suite 302, Boston, MA 02114, USA
| | - Arvin Halim
- Renal Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02114, USA; (A.H.)
| | - Tzong-shi Lu
- Renal Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02114, USA; (A.H.)
| | - Alan Ashworth
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (UCSF), 1450 3rd St, San Francisco, CA 94158, USA;
| | - Irene Chong
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Chester Beatty Laboratories, 237 Fulham Road, London SW3 6JB, UK;
| |
Collapse
|