1
|
Grigore TV, Leusink QM, Zuidscherwoude M, Bos C, Olauson H, Hoenderop J. Partial renal deletion of Klotho is not sufficient to impact renal electrolyte handling in distal convoluted tubule specific knock-out mice. Physiol Rep 2025; 13:e70297. [PMID: 40165603 PMCID: PMC11959153 DOI: 10.14814/phy2.70297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
Klotho controls renal electrolyte handling by modulating tubular reabsorption of calcium and phosphate through the epithelial calcium channel TRPV5 and sodium phosphate co-transporter NPT2A. The Ksp-KL-/- mice have a targeted deletion of Klotho in the distal part of the nephron. Considering that the distal convoluted tubule is the most important site for Ca2+ regulation in the kidney, Ksp-KL-/- mice were challenged with a Ca2+-deficient diet for determining the Ca2+ handling and pinpointing the Klotho levels needed for controlling renal Ca2+ handling. The Ksp-KL-/- mice displayed normal weight and showed unaltered calcium and phosphate levels in serum and 24-h urine. Expression of calciotropic (Trpv5, Trpv6) and phosphotropic (Slc34a1, Slc34a2) genes in the kidneys, duodenum, ileum, and colon were not affected by Klotho deletion. In conclusion, our study reports that mice with 18%-93% residual levels of Klotho in the kidney exhibit normal electrolyte homeostasis when placed on a low Ca2+-content diet.
Collapse
Affiliation(s)
- Teodora V. Grigore
- Department of Medical BiosciencesResearch Institute for Medical Innovation, Radboud University Medical CenterNijmegenThe Netherlands
| | - Quinty M. Leusink
- Department of Medical BiosciencesResearch Institute for Medical Innovation, Radboud University Medical CenterNijmegenThe Netherlands
| | - Malou Zuidscherwoude
- Department of Medical BiosciencesResearch Institute for Medical Innovation, Radboud University Medical CenterNijmegenThe Netherlands
| | - Caro Bos
- Department of Medical BiosciencesResearch Institute for Medical Innovation, Radboud University Medical CenterNijmegenThe Netherlands
| | - Hannes Olauson
- Division of Renal MedicineDepartment of Clinical Science, Intervention and Technology, Karolinska InstitutetStockholmSweden
| | - Joost Hoenderop
- Department of Medical BiosciencesResearch Institute for Medical Innovation, Radboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
2
|
Kamkari NA, Chen R, Bronson I, Coyne C. Acral Mesenchymal Tumor Leading to Tumor-Induced Osteomalacia: Case Report and Literature Review. AACE Clin Case Rep 2025; 11:143-147. [PMID: 40201463 PMCID: PMC11973645 DOI: 10.1016/j.aace.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/29/2024] [Accepted: 12/31/2024] [Indexed: 04/10/2025] Open
Abstract
Objective/Background Tumor-induced osteomalacia (TIO) is a rare paraneoplastic syndrome caused by excessive secretion of fibroblast growth factor 23 (FGF-23) by phosphaturic mesenchymal tumors. This leads to hypophosphatemia, vitamin D deficiency, and impaired bone metabolism. TIO is often misdiagnosed due to its rarity and nonspecific symptoms. Case Report We report a 58-year-old male presenting with multiple nontraumatic fractures, muscle weakness, and functional decline. Laboratory evaluation revealed hypophosphatemia, elevated parathyroid hormone, reduced 1,25-dihydroxyvitamin D, and markedly elevated FGF-23 levels. Imaging identified a soft tissue mass in the plantar region of the right foot, which was confirmed as a phosphaturic mesenchymal tumor upon pathological analysis. The patient underwent surgical resection, resulting in rapid normalization of biochemical abnormalities, including serum phosphorus, parathyroid hormone, and 1,25-dihydroxyvitamin D, within 5 days. Discussion This case underscores the importance of recognizing TIO in patients with unexplained hypophosphatemia and fractures. The curative potential of tumor resection was demonstrated with rapid biochemical and clinical improvement. Diagnostic challenges often arise due to the rarity and atypical presentation of these tumors, particularly in uncommon locations such as the plantar region. Emerging therapies, such as FGF-23 inhibitors like burosumab, provide alternatives for nonlocalizable or unresectable tumors. Conclusion This case emphasizes the need for increased clinician awareness, multidisciplinary approaches, and advances in diagnostic imaging to reduce delays in diagnosing TIO. Further research is necessary to elucidate the pathophysiology, explore genetic associations, and improve treatment options for this debilitating condition.
Collapse
Affiliation(s)
- Nick A. Kamkari
- University of Massachusetts Chan Medical School, Worcester, Massachusetts
| | - Ryan Chen
- University of Massachusetts Chan Medical School, Worcester, Massachusetts
| | - Isaac Bronson
- University of Massachusetts Chan Medical School, Worcester, Massachusetts
| | - Christopher Coyne
- University of Massachusetts Chan Medical School, Worcester, Massachusetts
| |
Collapse
|
3
|
Shahzamani K, Amooyi A, Karampoor S, Khanizadeh S, Farahmand M. Klotho protein: A key modulator of aging and COVID-19 severity. Int J Biol Macromol 2025; 296:139234. [PMID: 39798764 DOI: 10.1016/j.ijbiomac.2024.139234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 11/04/2024] [Accepted: 12/24/2024] [Indexed: 01/15/2025]
Abstract
The COVID-19 pandemic has drawn significant attention to factors affecting disease severity, especially in older adults. This study explores the relationship between Klotho, an anti-aging protein, and COVID-19 severity. Conducted at Tehran's Firouzgar Hospital, this case-control study involved 279 participants, assessing serum levels of Klotho, inflammatory markers (C-reactive protein (CRP), Interleukin 6 (IL-6)), and Vitamin D. The findings indicate significantly lower Klotho levels in COVID-19 patients, especially those in the ICU, which correlate with elevated inflammatory markers and reduced Vitamin D levels. This inverse relationship between Klotho levels and disease severity underscores the protein's potential modulatory role in the inflammatory response to COVID-19. The study not only highlights the importance of Klotho as a biomarker for aging and disease severity but also suggests its potential therapeutic value in managing COVID-19, offering a novel perspective on targeting aging-related pathways to mitigate the impact of the disease. These insights open new avenues for research and intervention strategies to leverage anti-aging mechanisms to combat COVID-19 and potentially other age-related diseases.
Collapse
Affiliation(s)
- Kiana Shahzamani
- Lorestan University of Medical Sciences, Islamic Republic of Iran.
| | - Atefeh Amooyi
- Lorestan University of Medical Sciences, Islamic Republic of Iran
| | - Sajad Karampoor
- Iran University of Medical Sciences, Islamic Republic of Iran
| | | | | |
Collapse
|
4
|
Ariadel-Cobo DG, Estébanez B, González-Arnáiz E, García-Pérez MP, Rivera-Viloria M, Pintor de la Maza B, Barajas-Galindo DE, García-Sastre D, Ballesteros-Pomar MD, Cuevas MJ. Influence of Klotho Protein Levels in Obesity and Sarcopenia: A Systematic Review. Int J Mol Sci 2025; 26:1915. [PMID: 40076542 PMCID: PMC11900336 DOI: 10.3390/ijms26051915] [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: 12/31/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
The Klotho gene is recognized for its anti-aging properties. Its downregulation leads to aging-like phenotypes, whereas overexpression can extend lifespan. Klotho protein exists in three forms: α-klotho, β-klotho and γ-klotho. The α-klotho has two isoforms: a membrane-bound form, primarily in the kidney and brain, and a secreted klotho protein present in blood, urine, and cerebrospinal fluid. Klotho functions as a co-receptor for fibroblast growth factor-23 (FGF23), regulating phosphate metabolism. The membrane-bound form controls various ion channels and receptors, while the secreted form regulates endocrine FGFs, including FGF19 and FGF21. The interaction between β-klotho and FGF21 in muscle is critical in the development of sarcopenic obesity. This systematic review, registered in PROSPERO and conducted following PRISMA guidelines, evaluates klotho levels in individuals with obesity or sarcopenic obesity. The study includes overweight, obese, and sarcopenic obese adults compared to those with a normal body mass index. After reviewing 713 articles, 20 studies were selected, including observational, cross-sectional, cohort studies, and clinical trials. Significant associations between klotho levels and obesity, metabolic syndrome (MS), and cardiovascular risk were observed. Exercise and dietary interventions positively influenced klotho levels, which were linked to improved muscle strength and slower decline. Klotho is a potential biomarker for obesity, MS, and sarcopenic obesity. Further research is needed to explore its mechanisms and therapeutic potential.
Collapse
Affiliation(s)
- Diana G. Ariadel-Cobo
- Institute of Biomedicine (IBIOMED), University of León, 24071 León, Spain; (D.G.A.-C.); (B.E.); (E.G.-A.); (M.R.-V.)
- Department of Endocrinology and Nutrition, Complejo Asistencial Universitario de León (CAULE), 24071 León, Spain; (M.P.G.-P.); (B.P.d.l.M.); (D.E.B.-G.); (D.G.-S.)
| | - Brisamar Estébanez
- Institute of Biomedicine (IBIOMED), University of León, 24071 León, Spain; (D.G.A.-C.); (B.E.); (E.G.-A.); (M.R.-V.)
| | - Elena González-Arnáiz
- Institute of Biomedicine (IBIOMED), University of León, 24071 León, Spain; (D.G.A.-C.); (B.E.); (E.G.-A.); (M.R.-V.)
- Department of Endocrinology and Nutrition, Complejo Asistencial Universitario de León (CAULE), 24071 León, Spain; (M.P.G.-P.); (B.P.d.l.M.); (D.E.B.-G.); (D.G.-S.)
| | - María Pilar García-Pérez
- Department of Endocrinology and Nutrition, Complejo Asistencial Universitario de León (CAULE), 24071 León, Spain; (M.P.G.-P.); (B.P.d.l.M.); (D.E.B.-G.); (D.G.-S.)
| | - Marta Rivera-Viloria
- Institute of Biomedicine (IBIOMED), University of León, 24071 León, Spain; (D.G.A.-C.); (B.E.); (E.G.-A.); (M.R.-V.)
| | - Begoña Pintor de la Maza
- Department of Endocrinology and Nutrition, Complejo Asistencial Universitario de León (CAULE), 24071 León, Spain; (M.P.G.-P.); (B.P.d.l.M.); (D.E.B.-G.); (D.G.-S.)
| | - David Emilio Barajas-Galindo
- Department of Endocrinology and Nutrition, Complejo Asistencial Universitario de León (CAULE), 24071 León, Spain; (M.P.G.-P.); (B.P.d.l.M.); (D.E.B.-G.); (D.G.-S.)
| | - Diana García-Sastre
- Department of Endocrinology and Nutrition, Complejo Asistencial Universitario de León (CAULE), 24071 León, Spain; (M.P.G.-P.); (B.P.d.l.M.); (D.E.B.-G.); (D.G.-S.)
| | - María D. Ballesteros-Pomar
- Institute of Biomedicine (IBIOMED), University of León, 24071 León, Spain; (D.G.A.-C.); (B.E.); (E.G.-A.); (M.R.-V.)
- Department of Endocrinology and Nutrition, Complejo Asistencial Universitario de León (CAULE), 24071 León, Spain; (M.P.G.-P.); (B.P.d.l.M.); (D.E.B.-G.); (D.G.-S.)
| | - María J. Cuevas
- Institute of Biomedicine (IBIOMED), University of León, 24071 León, Spain; (D.G.A.-C.); (B.E.); (E.G.-A.); (M.R.-V.)
| |
Collapse
|
5
|
Schnicker NJ, Xu Z, Amir M, Gakhar L, Huang CL. Conformational landscape of soluble α-klotho revealed by cryogenic electron microscopy. Sci Rep 2025; 15:543. [PMID: 39747283 PMCID: PMC11696049 DOI: 10.1038/s41598-024-84246-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
α-Klotho (KLA) is a type-1 membranous protein that can associate with fibroblast growth factor receptor (FGFR) to form co-receptor for FGF23. The ectodomain of unassociated KLA is shed as soluble KLA (sKLA) to exert FGFR/FGF23-independent pleiotropic functions. The previously determined X-ray crystal structure of the extracellular region of sKLA in complex with FGF23 and FGFR1c suggests that sKLA functions solely as an on-demand coreceptor for FGF23. To understand the FGFR/FGF23-independent pleiotropic functions of sKLA, we investigated biophysical properties and structure of apo-sKLA. Single particle cryogenic electron microscopy (cryo-EM) revealed a 3.3 Å resolution structure of apo-sKLA that overlays well with its counterpart in the ternary complex with several distinct features. Compared to the ternary complex, the KL2 domain of apo-sKLA is more flexible. Three-dimensional variability analysis revealed that apo-sKLA adopts conformations with different KL1-KL2 interdomain bending and rotational angles. Mass photometry revealed that sKLA can form a stable structure with FGFR and/or FGF23 as well as sKLA dimer in solution. Cryo-EM supported the dimeric structure of sKLA. Recent studies revealed that FGF23 contains two KLA-binding sites. Our computational studies revealed that each site binds separate KLA in the dimer. The potential multiple forms and shapes of sKLA support its role as FGFR-independent hormone with pleiotropic functions. The ability of FGF23 to engage two KLA's simultaneously raises a potential new mechanism of action for FGF23-mediated signaling by the membranous klotho.
Collapse
Affiliation(s)
- Nicholas J Schnicker
- Protein and Crystallography Facility, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| | - Zhen Xu
- Protein and Crystallography Facility, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Mohammad Amir
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Lokesh Gakhar
- Protein and Crystallography Facility, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA, 52242, USA
| | - Chou-Long Huang
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| |
Collapse
|
6
|
Bala N, Rafay RH, Glover SC, Alli AA. Activity of Various Cathepsin Proteases and Enrichment of Klotho Protein in the Urine and Urinary Extracellular Vesicles After SARS-CoV-2 Infection. Viruses 2024; 17:25. [PMID: 39861814 PMCID: PMC11768607 DOI: 10.3390/v17010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/20/2024] [Accepted: 12/25/2024] [Indexed: 01/27/2025] Open
Abstract
Background: The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is responsible for causing the Coronavirus disease 2019 (COVID-19) outbreak. While mutations cause the emergence of new variants, the ancestral SARS-CoV-2 strain is unique among other strains. Methods: Various clinical parameters, the activity of cathepsin proteases, and the concentration of various proteins were measured in urine samples from COVID-19-negative participants and COVID-19-positive participants. Urinary extracellular vesicles (uEVs) were isolated from urine samples from the two groups and used for proteomic analysis and subsequent pathway analyses. Results: Activity levels of cathepsin S and L were greater in the urine of COVID-19-positive participants. The concentration of C-reactive protein, transmembrane serine protease 2, and klotho protein were significantly greater in the urine of COVID-19-positive participants. There was a greater amount of uEVs in the COVID-19 group and klotho protein was found to be enriched in uEVs from the COVID-19 group. Pathway analyses of the proteomics data showed most of the identified proteins were involved in signal transduction, stress response, protein metabolism, and transport. The identified proteins were predominantly associated with cellular membranes and with function of the cytoskeleton, enzyme regulation, and signal transduction. Conclusions: Taken together, our data identify novel urinary biomarkers that could be used to further investigate the long-term effects of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Niharika Bala
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida College of Medicine, Gainesville, FL 32608, USA; (N.B.); (R.H.R.)
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32608, USA
| | - Ramish H. Rafay
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida College of Medicine, Gainesville, FL 32608, USA; (N.B.); (R.H.R.)
| | - Sarah C. Glover
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA;
| | - Abdel A. Alli
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida College of Medicine, Gainesville, FL 32608, USA; (N.B.); (R.H.R.)
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32608, USA
| |
Collapse
|
7
|
Cararo-Lopes MM, Sadovnik R, Fu A, Suresh S, Gandu S, Firestein BL. Overexpression of α-Klotho isoforms promotes distinct Effects on BDNF-Induced Alterations in Dendritic Morphology. Mol Neurobiol 2024; 61:9155-9170. [PMID: 38589756 PMCID: PMC11496329 DOI: 10.1007/s12035-024-04171-y] [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: 11/16/2023] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
α-Klotho (α-Kl) is a modulator of aging, neuroprotection, and cognition. Transcription of the Klotho gene produces two splice variants-a membrane protein (mKl), which can be cleaved and released into the extracellular milieu, and a truncated secreted form (sKl). Despite mounting evidence supporting a role for α-Kl in brain function, the specific roles of α-Kl isoforms in neuronal development remain elusive. Here, we examined α-Kl protein levels in rat brain and observed region-specific expression in the adult that differs between isoforms. In the developing hippocampus, levels of isoforms decrease after the third postnatal week, marking the end of the critical period for development. We overexpressed α-Kl isoforms in primary cultures of rat cortical neurons and evaluated effects on brain-derived neurotrophic factor (BDNF) signaling. Overexpression of either isoform attenuated BDNF-mediated signaling and reduced intracellular Ca2+ levels, with mKl promoting a greater effect. mKl or sKl overexpression in hippocampal neurons resulted in a partially overlapping reduction in secondary dendrite branching. Moreover, mKl overexpression increased primary dendrite number. BDNF treatment of neurons overexpressing sKl resulted in a dendrite branching phenotype similar to control neurons. In neurons overexpressing mKl, BDNF treatment restored branching of secondary and higher order dendrites close, but not distal, to the soma. Taken together, the data presented support the idea that sKl and mKl play distinct roles in neuronal development, and specifically, in dendrite morphogenesis.
Collapse
Affiliation(s)
- Marina Minto Cararo-Lopes
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Cell and Developmental Biology Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Ratchell Sadovnik
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Allen Fu
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Shradha Suresh
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Neuroscience Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Srinivasa Gandu
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Cell and Developmental Biology Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
| |
Collapse
|
8
|
Grigore TV, Zuidscherwoude M, Olauson H, Hoenderop JG. Lessons from Klotho mouse models to understand mineral homeostasis. Acta Physiol (Oxf) 2024; 240:e14220. [PMID: 39176993 DOI: 10.1111/apha.14220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/30/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024]
Abstract
AIM Klotho, a key component of the endocrine fibroblast growth factor receptor-fibroblast growth factor axis, is a multi-functional protein that impacts renal electrolyte handling. The physiological significance of Klotho will be highlighted in the regulation of calcium, phosphate, and potassium metabolism. METHODS In this review, we compare several murine models with different renal targeted deletions of Klotho and the insights into the molecular and physiological function that these models offer. RESULTS In vivo, Klotho deficiency is associated with severely impaired mineral metabolism, with consequences on growth, longevity and disease development. Additionally, we explore the perspectives of Klotho in renal pathology and vascular events, as well as potential Klotho treatment options. CONCLUSION This comprehensive review emphasizes the use of Klotho to shed light on deciphering the renal molecular in vivo mechanisms in electrolyte handling, as well as novel therapeutic interventions.
Collapse
Affiliation(s)
- Teodora V Grigore
- Department of Medical BioSciences, Radboud Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Malou Zuidscherwoude
- Department of Medical BioSciences, Radboud Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hannes Olauson
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Joost G Hoenderop
- Department of Medical BioSciences, Radboud Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
9
|
Mahtani T, Sheth H, Smith LK, Benedict L, Brecier A, Ghasemlou N, Treanor B. The ion channel TRPV5 regulates B-cell signaling and activation. Front Immunol 2024; 15:1386719. [PMID: 38694510 PMCID: PMC11061418 DOI: 10.3389/fimmu.2024.1386719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 03/28/2024] [Indexed: 05/04/2024] Open
Abstract
Introduction B-cell activation triggers the release of endoplasmic reticulum calcium stores through the store-operated calcium entry (SOCE) pathway resulting in calcium influx by calcium release-activated calcium (CRAC) channels on the plasma membrane. B-cell-specific murine knockouts of SOCE do not impact humoral immunity suggesting that alternative channels may be important. Methods We identified a member of the calcium-permeable transient receptor potential (TRP) ion channel family, TRPV5, as a candidate channel expressed in B cells by a quantitative polymerase chain reaction (qPCR) screen. To further investigate the role of TRPV5 in B-cell responses, we generated a murine TRPV5 knockout (KO) by CRISPR-Cas9. Results We found TRPV5 polarized to B-cell receptor (BCR) clusters upon stimulation in a PI3K-RhoA-dependent manner. TRPV5 KO mice have normal B-cell development and mature B-cell numbers. Surprisingly, calcium influx upon BCR stimulation in primary TRPV5 KO B cells was not impaired; however, differential expression of other calcium-regulating proteins, such as ORAI1, may contribute to a compensatory mechanism for calcium signaling in these cells. We demonstrate that TRPV5 KO B cells have impaired spreading and contraction in response to membrane-bound antigen. Consistent with this, TRPV5 KO B cells have reduced BCR signaling measured through phospho-tyrosine residues. Lastly, we also found that TRPV5 is important for early T-dependent antigen specific responses post-immunization. Discussion Thus, our findings identify a role for TRPV5 in BCR signaling and B-cell activation.
Collapse
Affiliation(s)
- Trisha Mahtani
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Hena Sheth
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - L. K. Smith
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON, Canada
| | - Leshawn Benedict
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON, Canada
| | - Aurelie Brecier
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Nader Ghasemlou
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Bebhinn Treanor
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
10
|
Doupnik CA, Luer CA, Walsh CJ, Restivo J, Brick JX. Bioactive Properties of Venoms Isolated from Whiptail Stingrays and the Search for Molecular Mechanisms and Targets. Pharmaceuticals (Basel) 2024; 17:488. [PMID: 38675448 PMCID: PMC11053709 DOI: 10.3390/ph17040488] [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: 02/27/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
The venom-containing barb attached to their 'whip-like' tail provides stingrays a defensive mechanism for evading predators such as sharks. From human encounters, dermal stingray envenomation is characterized by intense pain often followed by tissue necrosis occurring over several days to weeks. The bioactive components in stingray venoms (SRVs) and their molecular targets and mechanisms that mediate these complex responses are not well understood. Given the utility of venom-derived proteins from other venomous species for biomedical and pharmaceutical applications, we set out to characterize the bioactivity of SRV extracts from three local species that belong to the Dasyatoidea 'whiptail' superfamily. Multiple cell-based assays were used to quantify and compare the in vitro effects of these SRVs on different cell lines. All three SRVs demonstrated concentration-dependent growth-inhibitory effects on three different human cell lines tested. In contrast, a mouse fibrosarcoma cell line was markedly resistant to all three SRVs, indicating the molecular target(s) for mediating the SRV effects are not expressed on these cells. The multifunctional SRV responses were characterized by an acute disruption of cell adhesion leading to apoptosis. These findings aim to guide future investigations of individual SRV proteins and their molecular targets for potential use in biomedical applications.
Collapse
Affiliation(s)
- Craig A. Doupnik
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Carl A. Luer
- Marine Biomedical Research Program, Mote Marine Laboratory, Sarasota, FL 34236, USA;
| | - Catherine J. Walsh
- Marine Immunology Program, Mote Marine Laboratory, Sarasota, FL 34236, USA; (C.J.W.); (J.R.)
| | - Jessica Restivo
- Marine Immunology Program, Mote Marine Laboratory, Sarasota, FL 34236, USA; (C.J.W.); (J.R.)
| | - Jacqueline Xinlan Brick
- Department of Biology, College of Arts & Sciences, Oberlin College and Conservatory, Oberlin, OH 44074, USA;
| |
Collapse
|
11
|
Schnicker NJ, Xu Z, Amir M, Gakhar L, Huang CL. Conformational landscape of soluble α-klotho revealed by cryogenic electron microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.02.583144. [PMID: 38496408 PMCID: PMC10942382 DOI: 10.1101/2024.03.02.583144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
α-Klotho (KLA) is a type-1 membranous protein that can associate with fibroblast growth factor receptor (FGFR) to form co-receptor for FGF23. The ectodomain of unassociated KLA is shed as soluble KLA (sKLA) to exert FGFR/FGF23-independent pleiotropic functions. The previously determined X-ray crystal structure of the extracellular region of sKLA in complex with FGF23 and FGFR1c suggests that sKLA functions solely as an on-demand coreceptor for FGF23. To understand the FGFR/FGF23-independent pleiotropic functions of sKLA, we investigated biophysical properties and structure of apo-sKLA. Mass photometry revealed that sKLA can form a stable structure with FGFR and/or FGF23 as well as sKLA dimer in solution. Single particle cryogenic electron microscopy (cryo-EM) supported the dimeric structure of sKLA. Cryo-EM further revealed a 3.3Å resolution structure of apo-sKLA that overlays well with its counterpart in the ternary complex with several distinct features. Compared to the ternary complex, the KL2 domain of apo-sKLA is more flexible. 3D variability analysis revealed that apo-sKLA adopts conformations with different KL1-KL2 interdomain bending and rotational angles. The potential multiple forms and shapes of sKLA support its role as FGFR-independent hormone with pleiotropic functions. A comprehensive understanding of the sKLA conformational landscape will provide the foundation for developing klotho-related therapies for diseases.
Collapse
Affiliation(s)
- Nicholas J. Schnicker
- Protein and Crystallography Facility, University of Iowa Carver College of Medicine, Iowa City, Iowa, 52242, USA
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, Iowa, 52242, USA
| | - Zhen Xu
- Protein and Crystallography Facility, University of Iowa Carver College of Medicine, Iowa City, Iowa, 52242, USA
| | - Mohammad Amir
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, 52242, USA
| | - Lokesh Gakhar
- Protein and Crystallography Facility, University of Iowa Carver College of Medicine, Iowa City, Iowa, 52242, USA
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Chou-Long Huang
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, 52242, USA
| |
Collapse
|
12
|
Fan Z, Wei X, Zhu X, Yang K, Tian L, Du Y, Yang L. Correlation between soluble klotho and chronic kidney disease-mineral and bone disorder in chronic kidney disease: a meta-analysis. Sci Rep 2024; 14:4477. [PMID: 38396063 PMCID: PMC10891172 DOI: 10.1038/s41598-024-54812-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
We conducted a systematic search across medical databases, including PubMed, Web of Science, EMBASE, and Cochrane Library, up to March 2023. A total of 1944 subjects or individuals from 17 studies were included in our final analysis. The correlation coefficient (r) between sKlotho and calcium was [0.14, (0.02, 0.26)], and a moderate heterogeneity was observed (I2 = 66%, P < 0.05). The correlation coefficient (r) between Klotho and serum phosphate was [- 0.21, (- 0.37, - 0.04)], with apparent heterogeneity (I2 = 84%, P < 0.05). The correlation coefficient (r) between sKlotho and parathyroid hormone and vascular calcification was [- 0.23,(- 0.29, - 0.17); - 0.15, (- 0.23, - 0.08)], with no significant heterogeneity among the studies. (I2 = 40%, P < 0.05; I2 = 30%, P < 0.05). A significant correlation exists between low sKlotho levels and an increased risk of CKD-MBD in patients with CKD. According to the findings, sKlotho may play a role in alleviating CKD-MBD by lowering phosphorus and parathyroid hormone levels, regulating calcium levels, and suppressing vascular calcification. As analysis showed that sKlotho has an important impact on the pathogenesis and progression of CKD-MBD in CKD patients. Nonetheless, further comprehensive and high-quality studies are needed to validate our conclusions.
Collapse
Affiliation(s)
- Zhongyu Fan
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Xuejiao Wei
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Xiaoyu Zhu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Kun Yang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Ling Tian
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Yujun Du
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China.
| | - Liming Yang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
13
|
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
|
14
|
Huang HW, Chen CC, Lin KI, Hsu TL, Wong CH. Single Site N-Glycosylation of B Cell Maturation Antigen (BCMA) Inhibits γ-Secretase-Mediated Shedding and Improves Surface Retention and Cell Survival. ACS Chem Biol 2024; 19:153-161. [PMID: 38085681 DOI: 10.1021/acschembio.3c00592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
B cell maturation antigen (BCMA), a member of the tumor necrosis factor receptor (TNFR) family, on the cell surface plays a key role in maintaining the survival of plasma cells and malignant as well as inflammatory accessory cells. Therefore, targeting BCMA or disrupting its interaction with ligands has been a potential approach to cancer therapy. BCMA contains a single N-glycosylation site, but the function of N-glycan on BCMA is not understood. Here, we found that the N-glycosylation of BCMA promoted its cell-surface retention while removing the N-glycan increased BCMA secretion through γ-secretase-mediated shedding. Addition of γ-secretase inhibitor prevented nonglycosylated BCMA from shedding and protected cells from dexamethasone and TRAIL-induced apoptosis.
Collapse
Affiliation(s)
- Han-Wen Huang
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Chen-Chun Chen
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Kuo-I Lin
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Tsui-Ling Hsu
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Chi-Huey Wong
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| |
Collapse
|
15
|
Abboud M, Merenbakh-Lamin K, Volkov H, Ben-Neriah S, Ligumsky H, Bronfeld S, Keren-Khadmy N, Giladi M, Shomron N, Wolf I, Rubinek T. Revealing the tumor suppressive sequence within KL1 domain of the hormone Klotho. Oncogene 2024; 43:354-362. [PMID: 38040805 DOI: 10.1038/s41388-023-02904-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 12/03/2023]
Abstract
Klotho, a 1012 amino acid transmembrane protein, is a potent tumor suppressor in different cancer types. Klotho is composed of two internal repeats KL1 and KL2, and the tumor suppressor activity is primarily attributed to the KL1 domain. Despite its significant role in regulating various cancer-related pathways, the precise mechanism underlying its tumor suppressor activity remains unresolved. In this study, we aimed to identify the sequence responsible for the tumor suppressor function of Klotho and gain insights into its mechanism of action. To accomplish this, we generated expression vectors of truncated KL1 at the C and N-terminal regions and evaluated their ability to inhibit the colony formation of several cancer cell lines. Our findings demonstrated that truncated KL1 1-340 (KL340) effectively inhibited colony formation similar to KL1, while truncated KL1 1-320 (KL320) lost this activity. Furthermore, this correlated with the inhibitory effect of KL1 and KL340 on the Wnt/β-catenin pathway, whereas KL320 had no effect. Transcriptomic analysis of MCF-7 cells expressing the constructs revealed enriched pathways associated with tumor suppressor activity in KL1 and KL340. Interestingly, the α-fold predictor tool highlighted distinct differences in the α and β sheets of the TIM barrel fold of the truncated Klotho constructs, adding to our understanding of their structural variations. In summary, this study identified the 340 N-terminal amino acids as the sequence that possesses Klotho's tumor suppressor activity and reveals a critical role in the 320-340 sequence for this function. It also provides a foundation for the development of Klotho-based therapeutic approaches for cancer treatment.
Collapse
Affiliation(s)
- Marana Abboud
- The Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | | | - Hadas Volkov
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Edmond J. Safra Center for Bioinformatics at Tel-Aviv University, Tel Aviv, Israel
| | - Shira Ben-Neriah
- The Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Hagai Ligumsky
- The Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Sarai Bronfeld
- The Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Noa Keren-Khadmy
- The Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Moshe Giladi
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Internal Medicine Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Noam Shomron
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Edmond J. Safra Center for Bioinformatics at Tel-Aviv University, Tel Aviv, Israel
| | - Ido Wolf
- The Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tami Rubinek
- The Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
16
|
Chi PJ, Lee CJ, Hung SY, Tsai JP, Liou HH. Urinary Klotho Excretion: A Key Regulator of Sodium Homeostasis in Chronic Kidney Disease Stage 2-4. Med Sci Monit Basic Res 2023; 29:e942097. [PMID: 37987256 PMCID: PMC10637118 DOI: 10.12659/msmbr.942097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/12/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Soluble alpha-klotho (klotho) is considered an important regulator of mineral homeostasis in patients with chronic kidney disease (CKD). Since the mineral transport proteins are located on the apical membrane of renal tubular cells, we hypothesized that urine klotho may also be involved in their homeostasis. We aimed to investigate the associations between serum and urine klotho and their impacts on mineral homeostasis in patients with stage 2 to 4 CKD. MATERIAL AND METHODS Serum, spot urine, and 24-h urine of klotho were measured by using enzyme-linked immunosorbent assay. Fractional excretion of sodium, potassium, calcium, phosphate, magnesium, and klotho were calculated. RESULTS A total of 53 patients with CKD stages 2 to 4 were enrolled in this cross-sectional study. The mean age was 71.1±10.5 years, and 68% were men. Linear regression analysis showed that serum log-transformed klotho was negatively associated with log-transformed fractional excretion of klotho (log-FEKlotho) (ß=-0.085, P=0.02), showing that urinary klotho excretion could negatively regulate serum klotho levels. Moreover, our multivariate stepwise regression showed log-fractional excretion of sodium was positively associated with log-FEKlotho (ß=0.138, P=0.032). This implied urinary klotho excretion positively regulated urinary sodium excretion. CONCLUSIONS Our study showed that urine klotho excretion resulted in decreased serum klotho levels and enhanced urinary sodium excretion in patients with CKD stages 2 to 4. In addition to serum klotho, we found, for the first time, that urine klotho also played a significant role in sodium homeostasis.
Collapse
Affiliation(s)
- Po-Jui Chi
- Division of Nephrology, Department of Internal Medicine, E-DA Hospital, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Chung-Jen Lee
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien, Taiwan
| | - Shih-Yuan Hung
- Division of Nephrology, Department of Internal Medicine, E-DA Hospital, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Jen-Pi Tsai
- Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi General Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| | - Hung-Hsiang Liou
- Division of Nephrology, Department of Internal Medicine, E-DA Hospital, Kaohsiung, Taiwan
| |
Collapse
|
17
|
Sun F, Liang P, Wang B, Liu W. The fibroblast growth factor-Klotho axis at molecular level. Open Life Sci 2023; 18:20220655. [PMID: 37941788 PMCID: PMC10628560 DOI: 10.1515/biol-2022-0655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 06/03/2023] [Accepted: 06/10/2023] [Indexed: 11/10/2023] Open
Abstract
Klotho is a recently discovered protein that has positive effects on all systems of the body, for example, regulating calcium and phosphorus metabolism, protecting nerves, delaying aging and so on. Fibroblast growth factors (FGFs) are a group of polypeptides that function throughout the body by binding with cell surface FGF receptors (FGFRs). Endocrine FGFs require Klotho as a co-receptor for FGFRs. There is increasing evidence that Klotho participates in calcium and phosphorus regulation and metabolic regulation via the FGF-Klotho axis. Moreover, soluble Klotho can function as a separate hormone to regulate homeostasis on various ion channels and carrier channels on the cell surface. This review mainly explains the molecular basis of the membrane signaling mechanism of Klotho.
Collapse
Affiliation(s)
- Fuqiang Sun
- School of Anesthesiology, Weifang Medical University, Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, Weifang261053, Shandong, China
| | - Panpan Liang
- School of Basic Medical Sciences, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Bo Wang
- School of Anesthesiology, Weifang Medical University, Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, Weifang261053, Shandong, China
| | - Wenbo Liu
- Central Laboratory of the First Affiliated Hospital, Weifang Medical University, Weifang261000, Shandong, China
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Fibroblast growth factor (FGF) 23 is a bone-derived hormone that regulates phosphate and vitamin D metabolism by targeting the kidney. When highly elevated, such as in chronic kidney disease (CKD), FGF23 can also target the heart and induce pathologic remodeling. Here we discuss the mechanisms that underlie the physiologic and pathologic actions of FGF23, with focus on its FGF receptors (FGFR) and co-receptors. RECENT FINDINGS Klotho is a transmembrane protein that acts as an FGFR co-receptor for FGF23 on physiologic target cells. Klotho also exists as a circulating variant, and recent studies suggested that soluble klotho (sKL) can mediate FGF23 effects in cells that do not express klotho. Furthermore, it has been assumed that the actions of FGF23 do not require heparan sulfate (HS), a proteoglycan that acts as a co-receptor for other FGF isoforms. However, recent studies revealed that HS can be part of the FGF23:FGFR signaling complex and modulate FGF23-induced effects. SUMMARY sKL and HS have appeared as circulating FGFR co-receptors that modulate the actions of FGF23. Experimental studies suggest that sKL protects from and HS accelerates CKD-associated heart injury. However, the in vivo relevance of these findings is still speculative.
Collapse
Affiliation(s)
- S Madison Thomas
- Division of Nephrology and Section of Mineral Metabolism, Department of Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | |
Collapse
|
19
|
Muromachi K, Nakano R, Fujita-Yoshigaki J, Sugiya H, Tani-Ishii N. BMP-1-induced GBA1 nuclear accumulation provokes CCN2 mRNA expression via importin-β-mediated nucleocytoplasmic pathway. J Cell Commun Signal 2023:10.1007/s12079-023-00740-3. [DOI: 10.1007/s12079-023-00740-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/13/2023] [Indexed: 03/29/2023] Open
|
20
|
Murali M, Murali VP, Joseph MM, Rajan S, Maiti KK. Elucidating cell surface glycan imbalance through SERS guided metabolic glycan labelling: An appraisal of metastatic potential in cancer cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2022; 234:112506. [PMID: 35785648 DOI: 10.1016/j.jphotobiol.2022.112506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/14/2022] [Accepted: 06/25/2022] [Indexed: 06/15/2023]
Abstract
The intrinsic complexities of cell-surface glycans impede tracking the metabolic changes in cells. By coupling metabolic glycan labelling (MGL) and surface-enhanced Raman scattering (SERS), we employed the MGL-SERS strategy to elucidate the differential glycosylation pattern in cancer cell lines. Herein, for the first time, we are reporting an N-alkyl derivative of glucosamine (GlcNPhAlk) as a glycan labelling precursor. The extent of labelling was assessed by utilizing Raman imaging and verified by complementary fluorescence and Western blot analysis. MGL-SERS technique was implemented for a comparative evaluation of cell surface glycan imbalance in different cancer cells wherein a linear relationship between glycan expression and metastatic potential was established. Further, the effect of sialyltransferase inhibitor, P-3Fax-Neu5Ac, on metabolic labelling of GlcNPhAlk proved the incorporation of GlcNPhAlk to the terminal glycans through the sialic acid biosynthetic pathway. Hence, this methodology unveils the phenomenon of metastatic progression in cancer cells with inherent glycosylation-related dysplasia.
Collapse
Affiliation(s)
- Madhukrishnan Murali
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR- National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram 695019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Vishnu Priya Murali
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR- National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram 695019, Kerala, India
| | - Manu M Joseph
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR- National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram 695019, Kerala, India
| | - Soumya Rajan
- Government College, Kasargod 671123, Kerala, India
| | - Kaustabh Kumar Maiti
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR- National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram 695019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
21
|
The role of α-klotho in human cancer: molecular and clinical aspects. Oncogene 2022; 41:4487-4497. [PMID: 36038662 DOI: 10.1038/s41388-022-02440-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 07/14/2022] [Accepted: 08/10/2022] [Indexed: 11/08/2022]
Abstract
Klotho is a well-established longevity hormone. Its most prominent function is the regulation of phosphate homeostasis. However, klotho possesses multiple pleiotropic activities, including inhibition of major signaling pathways, reducing oxidative stress and suppressing inflammation. These activities are tightly associated with cancer, and klotho was discovered as a universal tumor suppressor. We review here novel molecular aspects of klotho activity in cancer, focusing on its structure-function relationships and clinical aspects regarding its expression, blood levels, clinical risk, and prognostic value in the clinical setting. In addition, the potential benefit of klotho treatment combined with chemotherapy, biological therapy, or immunotherapy, are discussed. Finally, as klotho was shown in preclinical models to inhibit cancer development and growth, we discuss various approaches to developing klotho-based therapies.
Collapse
|
22
|
Yanucil C, Kentrup D, Campos I, Czaya B, Heitman K, Westbrook D, Osis G, Grabner A, Wende AR, Vallejo J, Wacker MJ, Navarro-Garcia JA, Ruiz-Hurtado G, Zhang F, Song Y, Linhardt RJ, White K, Kapiloff M, Faul C. Soluble α-klotho and heparin modulate the pathologic cardiac actions of fibroblast growth factor 23 in chronic kidney disease. Kidney Int 2022; 102:261-279. [PMID: 35513125 PMCID: PMC9329240 DOI: 10.1016/j.kint.2022.03.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 03/14/2022] [Accepted: 03/29/2022] [Indexed: 01/03/2023]
Abstract
Fibroblast growth factor (FGF) 23 is a phosphate-regulating hormone that is elevated in patients with chronic kidney disease and associated with cardiovascular mortality. Experimental studies showed that elevated FGF23 levels induce cardiac hypertrophy by targeting cardiac myocytes via FGF receptor isoform 4 (FGFR4). A recent structural analysis revealed that the complex of FGF23 and FGFR1, the physiologic FGF23 receptor in the kidney, includes soluble α-klotho (klotho) and heparin, which both act as co-factors for FGF23/FGFR1 signaling. Here, we investigated whether soluble klotho, a circulating protein with cardio-protective properties, and heparin, a factor that is routinely infused into patients with kidney failure during the hemodialysis procedure, regulate FGF23/FGFR4 signaling and effects in cardiac myocytes. We developed a plate-based binding assay to quantify affinities of specific FGF23/FGFR interactions and found that soluble klotho and heparin mediate FGF23 binding to distinct FGFR isoforms. Heparin specifically mediated FGF23 binding to FGFR4 and increased FGF23 stimulatory effects on hypertrophic growth and contractility in isolated cardiac myocytes. When repetitively injected into two different mouse models with elevated serum FGF23 levels, heparin aggravated cardiac hypertrophy. We also developed a novel procedure for the synthesis and purification of recombinant soluble klotho, which showed anti-hypertrophic effects in FGF23-treated cardiac myocytes. Thus, soluble klotho and heparin act as independent FGF23 co-receptors with opposite effects on the pathologic actions of FGF23, with soluble klotho reducing and heparin increasing FGF23-induced cardiac hypertrophy. Hence, whether heparin injections during hemodialysis in patients with extremely high serum FGF23 levels contribute to their high rates of cardiovascular events and mortality remains to be studied.
Collapse
Affiliation(s)
- Christopher Yanucil
- Division of Nephrology and Hypertension, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dominik Kentrup
- Division of Nephrology and Hypertension, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA.,Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL, USA
| | - Isaac Campos
- Division of Nephrology and Hypertension, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brian Czaya
- Division of Nephrology and Hypertension, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kylie Heitman
- Division of Nephrology and Hypertension, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - David Westbrook
- Division of Nephrology and Hypertension, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gunars Osis
- Division of Nephrology and Hypertension, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alexander Grabner
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Adam R. Wende
- Division of Molecular & Cellular Pathology, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Julian Vallejo
- Department of Molecular Biosciences, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Michael J. Wacker
- Department of Molecular Biosciences, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Jose Alberto Navarro-Garcia
- Cardiorenal Translational Laboratory, Institute of Research, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Gema Ruiz-Hurtado
- Cardiorenal Translational Laboratory, Institute of Research, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fuming Zhang
- Departments of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Yuefan Song
- Departments of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Robert J. Linhardt
- Departments of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.,Departments of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Kenneth White
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michael Kapiloff
- Departments of Ophthalmology and Medicine, Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA, USA
| | - Christian Faul
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
23
|
Hou Z, Ding Q, Li Y, Zhao Z, Yan F, Li Y, Wang X, Xu J, Chen W, Wu G, Ruan X, Zhao L. Intestinal epithelial β Klotho is a critical protective factor in alcohol-induced intestinal barrier dysfunction and liver injury. EBioMedicine 2022; 82:104181. [PMID: 35908416 PMCID: PMC9352463 DOI: 10.1016/j.ebiom.2022.104181] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/27/2022] [Accepted: 07/06/2022] [Indexed: 10/26/2022] Open
|
24
|
Hua F, Chen X. β-Klotho inhibits CSF-1 secretion and delays the development of endometrial cancer. Cell Cycle 2022; 21:2132-2144. [PMID: 35762530 DOI: 10.1080/15384101.2022.2092180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Senescent cells can drive tumors development by promoting chronic inflammation. There is a significant correlation between β-Klotho expression profiles and endometrial cancer (EC). However, how β-Klotho regulates the occurrence and development of uterine EC remains to be further studied. Our research found that compared with normal endometrial tissues, β-Klotho expression levels in EC tissues were significantly reduced; overexpression of β-Klotho significantly inhibited aging, proliferation and migration but promoted apoptosis of EC cells cultured in vitro. In normal endometrial cells, results confirmed that reduced levels of β-Klotho promoted CSF-1 secretion, and the migration ability of macrophages was significantly enhanced when co-cultured with normal endometrial cells. In contrast, the expression of CSF-1 was significantly reduced after overexpression of β-Klotho in EC cells, and the macrophage migration ability is significantly weakened when co-cultured with EC cells. Therefore, we believe that β-Klotho influences macrophage migration by regulating the expression of CSF-1, thereby interfering with the progression of EC. We investigated in depth the mechanism of β-Klotho regulating CSF-1 secretion and found that β-Klotho inhibits the phosphorylation of p65, which blocked the nuclear translocation of p65, thereby inhibiting the secretion of CSF-1 by EC cells. The above results indicate that β-Klotho-mediated inhibition of CSF-1 secretion reduces the migration of macrophages to tumor tissue and delays the progression of EC.
Collapse
Affiliation(s)
- Fu Hua
- Department of Gynecology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xiaogang Chen
- Department of Orthopedics, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| |
Collapse
|
25
|
Jena S, Sarangi P, Das UK, Lamare AA, Rattan R. Serum α- Klotho Protein Can Be an Independent Predictive Marker of Oxidative Stress (OS) and Declining Glomerular Function Rate in Chronic Kidney Disease (CKD) Patients. Cureus 2022; 14:e25759. [PMID: 35812534 PMCID: PMC9268485 DOI: 10.7759/cureus.25759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2022] [Indexed: 12/03/2022] Open
Abstract
Introduction Chronic kidney disease (CKD) has been recognized as a global health problem. Progression of CKD to advanced stages is associated with a significant increase in the generation of reactive oxygen species (ROS). An antiaging protein, α-Klotho, is found expressed in the distal convoluted tubules of the kidney where, predominantly, it works to increase calcium absorption and potassium excretion in distal tubule via N-linked glycans. The association of serum α-Klotho with oxidative stress, inflammation, and fibrosis, as seen in CKD, highlights its importance for studying disease prognosis with declining glomerular function rate (GFR). Material and methods This was a case-control study consisting of 90 subjects. Fifty diagnosed cases of CKD attending the department of nephrology, SCB Medical College, Cuttack, Odisha, were included, and 40 age and sex-matched healthy volunteers were taken as control. Serum α-Klotho levels were measured using enzyme-linked immunosorbent assay kits. Oxidative stress by estimating the total oxidant load by ferrous oxidation-xylenol orange version 2 (FOX2) method and the total antioxidant capacity of serum by the ferric reducing ability of plasma (FRAP) method. Estimation of the estimated glomerular filtration rate (eGFR) was done using the Cockcroft and Gault equation. Results Serum α-Klotho (ng/ml) was found to be 2.59±0.98 in cases as compared to 0.24±0.09 in controls (p< 0.01). The serum total oxidant load (ng/ml) was 1.96±1.01 and 0.05±0.02 in cases and controls, respectively. Serum total antioxidant capacity (µM) was measured as 281.80±78.0 in cases and 862.82±51.86 in controls. (p< 0.01). Serum Klotho has a negative correlation with eGFR in CKD patients (r = -0.065; p = 0.648). Conclusion The serum α-Klotho level was significantly higher in CKD patients than in healthy volunteers. Both serum α-Klotho and oxidative stress were negatively correlated with eGFR in CKD patients. Serum α-Klotho can be a suitable biomarker in CKD patients with declining GFR.
Collapse
|
26
|
Alves AWDS, Sousa BL, Moura LFWG, Rebouças EDL, Coutinho MR, Silva AW, Chaves RP, Carneiro RF, Bezerra EHS, Guedes MIF, Florean EOPT, Nagano CS, Sampaio AH, Rocha BAM. Codium isthmocladum lectin 1 (CiL-1): Interaction with N-glycans explains antinociceptive and anti-inflammatory activities in adult zebrafish (Danio rerio). Int J Biol Macromol 2022; 208:1082-1089. [PMID: 35378162 DOI: 10.1016/j.ijbiomac.2022.03.209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/20/2022] [Accepted: 03/30/2022] [Indexed: 12/30/2022]
Abstract
Inflammation and oxidative stress are processes associated with different human diseases. They are treated using drugs that have several side effects. Seaweed are sources of potentially relevant natural compounds for use as treatment of these disorders. Lectins are able to reversibly interact with complex carbohydrates and modulate cell membrane glycosylated receptors through this interaction. This study aimed to determine the antinociceptive and anti-inflammatory potential of CiL-1 in adult zebrafish by modulation of TRPA1 through lectin-glycan binding. Possible neuromodulation by TRPA1 channel was also evaluated by camphor pretreatment. CiL-1 was efficacious at all tested doses, revealing anti-nociceptive and anti-inflammatory effects in adult zebrafish. This galactose-binding lectin was also able to reduce the content of ROS in brain and liver. In silico analyses showed CiL-1 interactions with both ligands tested. LacNac2 presents the most favorable binding energy with the protein. The interaction occurs at 4 subsites as an extended conformation at the site. LacNac2-Sia had a less favorable curved-shape interaction energy. Based on the predictions made for the oligosaccharides, a tetra-antenate putative glycan was schematically constructed, illustrating an interaction between TRPA1 N-glycan and CiL-1. This binding seems to be related to CiL-1 anti-inflammatory activity as result of receptor modulation.
Collapse
Affiliation(s)
- Antônio Willame da Silva Alves
- Laboratório de Biocristalografia - LABIC, Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, Campus do Pici s/n, bloco 907, Av. Mister Hull, Fortaleza, Ceará 60440-970, Brazil
| | - Bruno Lopes Sousa
- Faculdade de Filosofia Dom Aureliano Matos, Universidade Estadual do Ceará, Av. Dom Aureliano Matos, 2060, Limoeiro do Norte, Ceará 62930-000, Brazil
| | - Luiz Francisco Wemmenson Gonçalves Moura
- Laboratório de Biotecnologia e Biologia Molecular - LBBM, Centro de Ciências da Saúde, Universidade Estadual do Ceará, Campus do Itaperi, Fortaleza, Ceará, Brazil
| | - Emanuela de Lima Rebouças
- Laboratório de Biotecnologia e Biologia Molecular - LBBM, Centro de Ciências da Saúde, Universidade Estadual do Ceará, Campus do Itaperi, Fortaleza, Ceará, Brazil
| | - Marnielle Rodrigues Coutinho
- Laboratório de Biotecnologia e Biologia Molecular - LBBM, Centro de Ciências da Saúde, Universidade Estadual do Ceará, Campus do Itaperi, Fortaleza, Ceará, Brazil
| | - Antônio Wlisses Silva
- Laboratório de Biotecnologia e Biologia Molecular - LBBM, Centro de Ciências da Saúde, Universidade Estadual do Ceará, Campus do Itaperi, Fortaleza, Ceará, Brazil
| | - Renata Pinheiro Chaves
- Laboratório de Biotecnologia Marinha - BioMar-Lab, Departamento de Engenharia de Pesca, Universidade Federal do Ceará, Campus do Pici, Fortaleza, Ceará, Brazil
| | - Rômulo Farias Carneiro
- Laboratório de Biotecnologia Marinha - BioMar-Lab, Departamento de Engenharia de Pesca, Universidade Federal do Ceará, Campus do Pici, Fortaleza, Ceará, Brazil
| | - Eduardo Henrique Salviano Bezerra
- Laboratório de Biocristalografia - LABIC, Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, Campus do Pici s/n, bloco 907, Av. Mister Hull, Fortaleza, Ceará 60440-970, Brazil; Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais, Rua Giuseppe Máximo Scolfaro, Cidade Universitária, Campinas, São Paulo, Brazil
| | - Maria Izabel Florindo Guedes
- Laboratório de Biotecnologia e Biologia Molecular - LBBM, Centro de Ciências da Saúde, Universidade Estadual do Ceará, Campus do Itaperi, Fortaleza, Ceará, Brazil
| | | | - Celso Shiniti Nagano
- Laboratório de Biotecnologia Marinha - BioMar-Lab, Departamento de Engenharia de Pesca, Universidade Federal do Ceará, Campus do Pici, Fortaleza, Ceará, Brazil
| | - Alexandre Holanda Sampaio
- Laboratório de Biotecnologia Marinha - BioMar-Lab, Departamento de Engenharia de Pesca, Universidade Federal do Ceará, Campus do Pici, Fortaleza, Ceará, Brazil
| | - Bruno Anderson Matias Rocha
- Laboratório de Biocristalografia - LABIC, Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, Campus do Pici s/n, bloco 907, Av. Mister Hull, Fortaleza, Ceará 60440-970, Brazil.
| |
Collapse
|
27
|
Regulation of Aging and Longevity by Ion Channels and Transporters. Cells 2022; 11:cells11071180. [PMID: 35406743 PMCID: PMC8997527 DOI: 10.3390/cells11071180] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 12/10/2022] Open
Abstract
Despite significant advances in our understanding of the mechanisms that underlie age-related physiological decline, our ability to translate these insights into actionable strategies to extend human healthspan has been limited. One of the major reasons for the existence of this barrier is that with a few important exceptions, many of the proteins that mediate aging have proven to be undruggable. The argument put forth here is that the amenability of ion channels and transporters to pharmacological manipulation could be leveraged to develop novel therapeutic strategies to combat aging. This review delves into the established roles for ion channels and transporters in the regulation of aging and longevity via their influence on membrane excitability, Ca2+ homeostasis, mitochondrial and endolysosomal function, and the transduction of sensory stimuli. The goal is to provide the reader with an understanding of emergent themes, and prompt further investigation into how the activities of ion channels and transporters sculpt the trajectories of cellular and organismal aging.
Collapse
|
28
|
van Megen WH, Beggs MR, An SW, Ferreira PG, Lee JJ, Wolf MT, Alexander RT, Dimke H. Gentamicin Inhibits Ca 2+ Channel TRPV5 and Induces Calciuresis Independent of the Calcium-Sensing Receptor-Claudin-14 Pathway. J Am Soc Nephrol 2022; 33:547-564. [PMID: 35022312 PMCID: PMC8975070 DOI: 10.1681/asn.2021030392] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 12/19/2021] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Treatment with the aminoglycoside antibiotic gentamicin can be associated with severe adverse effects, including renal Ca2+ wasting. The underlying mechanism is unknown but it has been proposed to involve activation of the Ca2+-sensing receptor (CaSR) in the thick ascending limb, which would increase expression of claudin-14 (CLDN14) and limit Ca2+ reabsorption. However, no direct evidence for this hypothesis has been presented. METHODS We studied the effect of gentamicin in vivo using mouse models with impaired Ca2+ reabsorption in the proximal tubule and the thick ascending limb. We used a Cldn14 promoter luciferase reporter assay to study CaSR activation and investigated the effect of gentamicin on activity of the distal nephron Ca2+ channel transient receptor potential vanilloid 5 (TRPV5), as determined by patch clamp in HEK293 cells. RESULTS Gentamicin increased urinary Ca2+ excretion in wild-type mice after acute and chronic administration. This calciuretic effect was unaltered in mice with genetic CaSR overactivation and was present in furosemide-treated animals, whereas the calciuretic effect in Cldn14-/- mice and mice with impaired proximal tubular Ca2+ reabsorption (claudin-2 [CLDN2]-deficient Cldn2-/- mice) was equivalent to that of wild-type mice. In vitro, gentamicin failed to activate the CaSR. In contrast, patch clamp analysis revealed that gentamicin strongly inhibited rabbit and human TRPV5 activity and chronic gentamicin administration downregulated distal nephron Ca2+ transporters. CONCLUSIONS Gentamicin does not cause hypercalciuria via activation of the CaSR-CLDN14 pathway or by interfering with proximal tubular CLDN2-dependent Ca2+ reabsorption. Instead, gentamicin blocks distal Ca2+ reabsorption by direct inhibition of the Ca2+ channel TRPV5. These findings offer new insights into Ca2+ wasting in patients treated with gentamicin.
Collapse
Affiliation(s)
- Wouter H. van Megen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Megan R. Beggs
- Department of Physiology, University of Alberta, Canada,Women and Children's Health Institute, Alberta, Canada
| | - Sung-Wan An
- Department of Pediatrics, Division of Pediatric Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Patrícia G. Ferreira
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Justin J. Lee
- Department of Physiology, University of Alberta, Canada
| | - Matthias T. Wolf
- Department of Pediatrics, Division of Pediatric Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - R. Todd Alexander
- Department of Physiology, University of Alberta, Canada,Women and Children's Health Institute, Alberta, Canada,Department of Pediatrics, University of Alberta, Canada
| | - Henrik Dimke
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark .,Department of Nephrology, Odense University Hospital, Denmark
| |
Collapse
|
29
|
S-Klotho level and physiological markers of cardiometabolic risk in healthy adult men. Aging (Albany NY) 2022; 14:708-727. [PMID: 35093938 PMCID: PMC8833136 DOI: 10.18632/aging.203861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 01/17/2022] [Indexed: 11/25/2022]
Abstract
S-Klotho is perceived as a biomarker of healthy aging that has been shown to be inversely associated with cardiometabolic risk in elderly individuals. The aim of this study was to test if s-Klotho level is associated with cardiometabolic risk markers in younger healthy men in order to verify the possible role of s-Klotho level as an early marker of cardiometabolic risk. A cross-sectional study was conducted among 186 healthy men (Mage=35.33, SDage=3.47) from a Western urban population. Serum basal levels of s-Klotho, lipid profile, homocysteine, glycemia markers, C-reactive protein, liver transaminases and creatinine were evaluated. Also, blood pressure was measured and cardiometabolic risk score and homeostatic model assessment for insulin resistance (HOMA-IR) were calculated. Testosterone and cortisol levels, self-reported psychological stress, physical activity, smoking in the past, alcohol use and body adiposity were controlled for. We found no relationship between levels of s-Klotho and physiological markers of cardiometabolic risk in the studied population. The results were similar when controlled for adiposity, testosterone level, physical activity, alcohol use and smoking in the past. We suggest that s-Klotho level is not an early marker of cardiometabolic risk in younger middle-aged healthy men.
Collapse
|
30
|
Franco ML, Beyerstedt S, Rangel ÉB. Klotho and Mesenchymal Stem Cells: A Review on Cell and Gene Therapy for Chronic Kidney Disease and Acute Kidney Disease. Pharmaceutics 2021; 14:11. [PMID: 35056905 PMCID: PMC8778857 DOI: 10.3390/pharmaceutics14010011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease (CKD) and acute kidney injury (AKI) are public health problems, and their prevalence rates have increased with the aging of the population. They are associated with the presence of comorbidities, in particular diabetes mellitus and hypertension, resulting in a high financial burden for the health system. Studies have indicated Klotho as a promising therapeutic approach for these conditions. Klotho reduces inflammation, oxidative stress and fibrosis and counter-regulates the renin-angiotensin-aldosterone system. In CKD and AKI, Klotho expression is downregulated from early stages and correlates with disease progression. Therefore, the restoration of its levels, through exogenous or endogenous pathways, has renoprotective effects. An important strategy for administering Klotho is through mesenchymal stem cells (MSCs). In summary, this review comprises in vitro and in vivo studies on the therapeutic potential of Klotho for the treatment of CKD and AKI through the administration of MSCs.
Collapse
Affiliation(s)
- Marcella Liciani Franco
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil; (M.L.F.); (S.B.)
| | - Stephany Beyerstedt
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil; (M.L.F.); (S.B.)
| | - Érika Bevilaqua Rangel
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil; (M.L.F.); (S.B.)
- Nephrology Division, Federal University of São Paulo, Sao Paulo 04038-901, Brazil
| |
Collapse
|
31
|
Arbel Rubinstein T, Reuveni I, Hesin A, Klein-Goldberg A, Olauson H, Larsson TE, Abraham CR, Zeldich E, Bosch A, Chillón M, Hollander KS, Shabtay-Orbach A, Vainer GW, Wolf I, Rubinek T. A Transgenic Model Reveals the Role of Klotho in Pancreatic Cancer Development and Paves the Way for New Klotho-Based Therapy. Cancers (Basel) 2021; 13:cancers13246297. [PMID: 34944918 PMCID: PMC8699737 DOI: 10.3390/cancers13246297] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/05/2021] [Accepted: 12/13/2021] [Indexed: 11/20/2022] Open
Abstract
Simple Summary We aimed to study the role of the anti-aging protein klotho and its secreted isoform, sKL, in pancreatic cancer. Three in vivo models, including a novel genetic mouse model and bioinformatics analyses, indicated klotho as a tumor suppressor in pancreatic ductal adenocarcinoma, and unveiled a unique klotho DNA hypermethylation pattern in pancreatic tumors. These results possess significant prognostic value, and further suggest that sKL may serve as a therapeutic agent for pancreatic ductal adenocarcinoma. Abstract Klotho is an anti-aging transmembrane protein, which can be shed and can function as a hormone. Accumulating data indicate that klotho is a tumor suppressor in a wide array of malignancies, and designate the subdomain KL1 as the active region of the protein towards this activity. We aimed to study the role of klotho as a tumor suppressor in pancreatic ductal adenocarcinoma (PDAC). Bioinformatics analyses of The Cancer Genome Atlas (TCGA) datasets revealed a correlation between the survival of PDAC patients, levels of klotho expression, and DNA methylation, and demonstrated a unique hypermethylation pattern of klotho in pancreatic tumors. The in vivo effects of klotho and KL1 were examined using three mouse models. Employing a novel genetic model, combining pancreatic klotho knockdown with a mutation in Kras, the lack of klotho contributed to PDAC generation and decreased mousece survival. In a xenograft model, administration of viral particles carrying sKL, a spliced klotho isoform containing the KL1 domain, inhibited pancreatic tumors. Lastly, treatment with soluble sKL prolonged survival of Pdx1-Cre; KrasG12D/+;Trp53R172H/+ (KPC) mice, a model known to recapitulate human PDAC. In conclusion, this study provides evidence that klotho is a tumor suppressor in PDAC. Furthermore, these data suggest that the levels of klotho expression and DNA methylation could have prognostic value in PDAC patients, and that administration of exogenous sKL may serve as a novel therapeutic strategy to treat PDAC.
Collapse
Affiliation(s)
- Tammi Arbel Rubinstein
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (T.A.R.); (I.R.); (A.H.); (A.K.-G.); (K.S.H.); (A.S.-O.); (I.W.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Inbal Reuveni
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (T.A.R.); (I.R.); (A.H.); (A.K.-G.); (K.S.H.); (A.S.-O.); (I.W.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Arkadi Hesin
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (T.A.R.); (I.R.); (A.H.); (A.K.-G.); (K.S.H.); (A.S.-O.); (I.W.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Anat Klein-Goldberg
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (T.A.R.); (I.R.); (A.H.); (A.K.-G.); (K.S.H.); (A.S.-O.); (I.W.)
| | - Hannes Olauson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14186 Stockholm, Sweden; (H.O.); (T.E.L.)
| | - Tobias E. Larsson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14186 Stockholm, Sweden; (H.O.); (T.E.L.)
- Department of Nephrology, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Carmela R. Abraham
- Boston University School of Medicine, Department of Biochemistry, Boston, MA 02118, USA; (C.R.A.); (E.Z.)
- Pharmacology & Experimental Therapeutics, Boston, MA 02118, USA
- Klogenix Therapeutics Inc., Boston, MA 02116, USA; (A.B.); (M.C.)
| | - Ella Zeldich
- Boston University School of Medicine, Department of Biochemistry, Boston, MA 02118, USA; (C.R.A.); (E.Z.)
- Klogenix Therapeutics Inc., Boston, MA 02116, USA; (A.B.); (M.C.)
| | - Assumpció Bosch
- Klogenix Therapeutics Inc., Boston, MA 02116, USA; (A.B.); (M.C.)
- Institut de Neurociències, Department of Biochemistry and Molecular Biology, Universitat Autonoma de Barcelona, 08035 Barcelona, Spain
- Unitat Mixta UAB-VHIR, Vall d’Hebron Institut de Recerca (VHIR), 08035 Barcelona, Spain
- CIBERNED, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miguel Chillón
- Klogenix Therapeutics Inc., Boston, MA 02116, USA; (A.B.); (M.C.)
- Institut de Neurociències, Department of Biochemistry and Molecular Biology, Universitat Autonoma de Barcelona, 08035 Barcelona, Spain
- Unitat Mixta UAB-VHIR, Vall d’Hebron Institut de Recerca (VHIR), 08035 Barcelona, Spain
- ICREA, Institut Catalan Recerca Avançada, 08010 Barcelona, Spain
| | - Kenneth Samuel Hollander
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (T.A.R.); (I.R.); (A.H.); (A.K.-G.); (K.S.H.); (A.S.-O.); (I.W.)
| | - Ayelet Shabtay-Orbach
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (T.A.R.); (I.R.); (A.H.); (A.K.-G.); (K.S.H.); (A.S.-O.); (I.W.)
| | - Gilad W. Vainer
- Pathology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel;
| | - Ido Wolf
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (T.A.R.); (I.R.); (A.H.); (A.K.-G.); (K.S.H.); (A.S.-O.); (I.W.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tami Rubinek
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (T.A.R.); (I.R.); (A.H.); (A.K.-G.); (K.S.H.); (A.S.-O.); (I.W.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Correspondence: ; Tel.: +972-527466151
| |
Collapse
|
32
|
Khan EA, Cheddani L, Saint-Jacques C, Vargas-Poussou R, Frochot V, Chieze R, Letavernier E, Avellino V, Lionnet F, Haymann JP. Primary Hyperparathyroidism in Homozygous Sickle Cell Patients: A Hemolysis-Mediated Hypocalciuric Hypercalcemia Phenotype? J Clin Med 2021; 10:jcm10215179. [PMID: 34768698 PMCID: PMC8584729 DOI: 10.3390/jcm10215179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/21/2021] [Accepted: 11/03/2021] [Indexed: 11/16/2022] Open
Abstract
Primary hyperparathyroidism (pHPT) has been reported to have a higher prevalence in sickle cell disease (SCD) patients, including a high rate of recurrence following surgery. However, most patients are asymptomatic at the time of diagnosis, with surprisingly infrequent hypercalciuria, raising the issue of renal calcium handling in SCD patients. We conducted a retrospective study including (1) 64 hypercalcemic pHPT non-SCD patients; (2) 177 SCD patients, divided into two groups of 12 hypercalcemic pHPT and 165 non-pHPT; (3) eight patients with a diagnosis of familial hypocalciuric hypercalcemia (FHH). Demographic and biological parameters at the time of diagnosis were collected and compared between the different groups. Determinants of fasting fractional excretion of calcium (FeCa2+) were also analyzed in non-pHPT SCD patients. Compared to non-SCD pHPT patients, our data show a similar ionized calcium and PTH concentration, with a lower plasmatic calcitriol concentration and a lower daily urinary calcium excretion in pHPT SCD patients (p < 0.0001 in both cases). Fasting FeCa2+ is also surprisingly low in pHPT SCD patients, and thus inadequate to be considered hypercalcemia, recalling the FHH phenotype. FeCa2+ is also low in the non-pHPT SCD control group, and negatively associated with PTH and hemolytic biomarkers such as LDH and low hemoglobin. Our data suggest that the pHPT biochemical phenotype in SCD patients resembles the FHH phenotype, and the fasting FeCa2+ association with chronic hemolysis biomarkers strengthens the view of a potential pharmacological link between hemolytic by-products and calcium reabsorption, potentially through a decreased calcium-sensing receptor (CaSR) activity.
Collapse
Affiliation(s)
- Edmat Akhtar Khan
- Service de Néphrologie, Université de Lorraine, CHRU-Nancy, 54500 Vandœuvre-lès-Nancy, France;
| | - Lynda Cheddani
- Unité HTA, Prévention et Thérapeutique Cardiovasculaires, Assistance Publique—Hôpitaux de Paris, Hôpital Hôtel Dieu, 75004 Paris, France;
- Centre de Diagnostic et de Thérapeutique, Hôtel-Dieu, Université de Paris, 75006 Paris, France
| | - Camille Saint-Jacques
- Service des Explorations Fonctionnelles Multidisciplinaires, Assistance Publique—Hôpitaux de Paris, Hôpital Tenon, 75020 Paris, France; (C.S.-J.); (V.F.); (R.C.); (E.L.)
- Unité Mixte de Recherche (UMR) S 1155, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Hôpital Tenon, 75020 Paris, France
| | - Rosa Vargas-Poussou
- Centre d’Investigation Clinique, Centre de Référence des Maladies Rénales Héréditaires de l’Enfant et de l’Adulte, Assistance Publique—Hôpitaux de Paris, Hôpital Européen Georges Pompidou, 75015 Paris, France;
| | - Vincent Frochot
- Service des Explorations Fonctionnelles Multidisciplinaires, Assistance Publique—Hôpitaux de Paris, Hôpital Tenon, 75020 Paris, France; (C.S.-J.); (V.F.); (R.C.); (E.L.)
- Unité Mixte de Recherche (UMR) S 1155, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Hôpital Tenon, 75020 Paris, France
| | - Remi Chieze
- Service des Explorations Fonctionnelles Multidisciplinaires, Assistance Publique—Hôpitaux de Paris, Hôpital Tenon, 75020 Paris, France; (C.S.-J.); (V.F.); (R.C.); (E.L.)
- Unité Mixte de Recherche (UMR) S 1155, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Hôpital Tenon, 75020 Paris, France
| | - Emmanuel Letavernier
- Service des Explorations Fonctionnelles Multidisciplinaires, Assistance Publique—Hôpitaux de Paris, Hôpital Tenon, 75020 Paris, France; (C.S.-J.); (V.F.); (R.C.); (E.L.)
- Unité Mixte de Recherche (UMR) S 1155, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Hôpital Tenon, 75020 Paris, France
| | - Virginie Avellino
- Service de Médecine Interne, Centre de Référence de la Drépanocytose, Assistance Publique—Hôpitaux de Paris, Hôpital Tenon, 75020 Paris, France; (V.A.); (F.L.)
| | - Francois Lionnet
- Service de Médecine Interne, Centre de Référence de la Drépanocytose, Assistance Publique—Hôpitaux de Paris, Hôpital Tenon, 75020 Paris, France; (V.A.); (F.L.)
| | - Jean-Philippe Haymann
- Service des Explorations Fonctionnelles Multidisciplinaires, Assistance Publique—Hôpitaux de Paris, Hôpital Tenon, 75020 Paris, France; (C.S.-J.); (V.F.); (R.C.); (E.L.)
- Unité Mixte de Recherche (UMR) S 1155, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Hôpital Tenon, 75020 Paris, France
- Correspondence: ; Tel.: +33-1-5601-6771; Fax: +33-1-5601-7003
| |
Collapse
|
33
|
Živanović J, Jarić I, Ajdžanović V, Miler M, Stanković S, Milošević V, Filipović B. Genistein regulates calcium and phosphate homeostasis without activation of MEK 1/2 signalling pathway in an animal model of the andropause. Ann Anat 2021; 239:151836. [PMID: 34563672 DOI: 10.1016/j.aanat.2021.151836] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 01/02/2023]
Abstract
Soy isoflavone genistein interplays with numerous physiological or pathophysiological processes during ageing. However, its protective role and underlying mechanisms of action in the regulation of calcium (Ca2+) and phosphate (Pi) homeostasis in an animal model of the andropause are yet to be fully clarified. Wistar male rats (16-month-old) were divided into sham-operated, orchidectomized, orchidectomized estradiol-treated (0.625 mg/kg b.m./day) and orchidectomized genistein-treated (30 mg/kg b.m./day) groups. Treatments were administered subcutaneously for 3 weeks, while the controls received vehicle alone. Estradiol treatment increased the expression level of fibroblast growth factor receptor (FGFR) and parathyroid hormone 1 receptor (PTH1R), and activated mitogen - activated protein kinase kinase 1/2 (MEK 1/2) signaling pathway in the kidneys. Genistein application induced a prominent gene and protein expression of Klotho and downregulated the expression of FGFR and PTH1R in the kidney of andropausal rats. Activation of protein kinase B (Akt) signalling pathway was observed, while MEK 1/2 signaling pathway wasn't altered after genistein treatment. The increase of 25 (OH) vitamin D in the serum and decrease in Ca2+ urine content was observed after genistein application. Our findings strongly suggest genistein as a potent biocompound with beneficial effects on the regulation of Ca2+ and Pi homeostasis, especially during aging process when the balance of mineral metabolism is impaired. These novel data provide closer insights into the physiological roles of genistein in the regulation of mineral homeostasis.
Collapse
Affiliation(s)
- Jasmina Živanović
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| | - Ivana Jarić
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia; Animal Welfare Division, Vetsuisse, University of Bern, Bern, Switzerland
| | - Vladimir Ajdžanović
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Marko Miler
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Sanja Stanković
- Center for Medical Biochemistry, Clinical Centre of Serbia, Belgrade, Serbia
| | - Verica Milošević
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Branko Filipović
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
34
|
Kuro-o M. Klotho and calciprotein particles as therapeutic targets against accelerated ageing. Clin Sci (Lond) 2021; 135:1915-1927. [PMID: 34374422 PMCID: PMC8355631 DOI: 10.1042/cs20201453] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 01/25/2023]
Abstract
The klotho gene, named after a Greek goddess who spins the thread of life, was identified as a putative 'ageing-suppressor' gene. Klotho-deficient mice exhibit complex ageing-like phenotypes including hypogonadism, arteriosclerosis (vascular calcification), cardiac hypertrophy, osteopenia, sarcopenia, frailty, and premature death. Klotho protein functions as the obligate co-receptor for fibroblast growth factor-23 (FGF23), a bone-derived hormone that promotes urinary phosphate excretion in response to phosphate intake. Thus, Klotho-deficient mice suffer not only from accelerated ageing but also from phosphate retention due to impaired phosphate excretion. Importantly, restoration of the phosphate balance by placing Klotho-deficient mice on low phosphate diet rescued them from premature ageing, leading us to the notion that phosphate accelerates ageing. Because the extracellular fluid is super-saturated in terms of phosphate and calcium ions, an increase in the phosphate concentration can trigger precipitation of calcium-phosphate. In the blood, calcium-phosphate precipitated upon increase in the blood phosphate concentration is adsorbed by serum protein fetuin-A to form colloidal nanoparticles called calciprotein particles (CPPs). In the urine, CPPs appear in the renal tubular fluid when FGF23 increases phosphate load excreted per nephron. CPPs can induce cell damage, ectopic calcification, and inflammatory responses. CPPs in the blood can induce arteriosclerosis and non-infectious chronic inflammation, whereas CPPs in the urine can induce renal tubular damage and interstitial inflammation/fibrosis. Thus, we propose that CPPs behave like a pathogen that accelerates ageing and should be regarded as a novel therapeutic target against age-related disorders including chronic kidney disease.
Collapse
Affiliation(s)
- Makoto Kuro-o
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| |
Collapse
|
35
|
Tang PK, Geddes RF, Jepson RE, Elliott J. A feline-focused review of chronic kidney disease-mineral and bone disorders - Part 1: Physiology of calcium handling. Vet J 2021; 275:105719. [PMID: 34311095 DOI: 10.1016/j.tvjl.2021.105719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 06/03/2021] [Accepted: 07/21/2021] [Indexed: 01/01/2023]
Abstract
Mineral derangements are a common consequence of chronic kidney disease (CKD). Despite the well-established role of phosphorus in the pathophysiology of CKD, the implications of calcium disturbances associated with CKD remain equivocal. Calcium plays an essential role in numerous physiological functions in the body and is a fundamental structural component of bone. An understanding of calcium metabolism is required to understand the potential adverse clinical implications and outcomes secondary to the (mal)adaptation of calcium-regulating hormones in CKD. The first part of this two-part review covers the physiology of calcium homeostasis (kidneys, intestines and bones) and details the intimate relationships between calcium-regulating hormones (parathyroid hormone, calcitriol, fibroblast growth factor 23, α-Klotho and calcitonin) and the role of the calcium-sensing receptor.
Collapse
Affiliation(s)
- Pak-Kan Tang
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London, UK.
| | - Rebecca F Geddes
- Department of Clinical Science and Services, Royal Veterinary College, University of London, London, UK
| | - Rosanne E Jepson
- Department of Clinical Science and Services, Royal Veterinary College, University of London, London, UK
| | - Jonathan Elliott
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London, UK
| |
Collapse
|
36
|
Chen P, De Schutter K, Van Damme EJM, Smagghe G. Can Plant Lectins Help to Elucidate Insect Lectin-Mediated Immune Response? INSECTS 2021; 12:insects12060497. [PMID: 34071763 PMCID: PMC8226959 DOI: 10.3390/insects12060497] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 12/25/2022]
Abstract
Simple Summary Lectins are proteins that can recognize and selectively bind specific sugar structures. These proteins are present in all kingdoms of life, including plants, animals, fungi and microorganisms and play a role in a broad range of processes. The interactions between lectins and their target carbohydrates play a primordial role in plant and animal immune systems. Despite being the largest and most diverse taxa on earth, the study of lectins and their functions in insects is lagging behind. To study the role of insect lectins in the immune response, plant lectins could provide an interesting tool. Plant lectins have been well characterized and many of them possess immunomodulatory properties in vertebrate cells. The increasing knowledge on the immunomodulatory effects of plant lectins could complement the missing knowledge on the endogenous insect lectins and contribute to understanding the processes and mechanisms by which lectins participate in insect immunity. This review summarizes existing studies of immune responses stimulated by endogenous or exogenous lectins. Abstract Lectins are carbohydrate-binding proteins that recognize and selectively bind to specific sugar structures. This group of proteins is widespread in plants, animals, and microorganisms, and exerts a broad range of functions. Many plant lectins were identified as exogenous stimuli of vertebrate immunity. Despite being the largest and most diverse taxon on earth, the study of lectins and their functions in insects is lagging behind. In insects, research on lectins and their biological importance has mainly focused on the C-type lectin (CTL) family, limiting our global understanding of the function of insect lectins and their role in insect immunity. In contrast, plant lectins have been well characterized and the immunomodulatory effects of several plant lectins have been documented extensively in vertebrates. This information could complement the missing knowledge on endogenous insect lectins and contribute to understanding of the processes and mechanisms by which lectins participate in insect immunity. This review summarizes existing studies of immune responses stimulated by endogenous or exogenous lectins. Understanding how lectins modulate insect immune responses can provide insight which, in turn, can help to elaborate novel ideas applicable for the protection of beneficial insects and the development of novel pest control strategies.
Collapse
Affiliation(s)
- Pengyu Chen
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (P.C.); (K.D.S.)
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium;
| | - Kristof De Schutter
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (P.C.); (K.D.S.)
| | - Els J. M. Van Damme
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium;
| | - Guy Smagghe
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (P.C.); (K.D.S.)
- Correspondence:
| |
Collapse
|
37
|
Tsuchiya K, Akihisa T. The Importance of Phosphate Control in Chronic Kidney Disease. Nutrients 2021; 13:nu13051670. [PMID: 34069053 PMCID: PMC8156430 DOI: 10.3390/nu13051670] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/06/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
A series of problems including osteopathy, abnormal serum data, and vascular calcification associated with chronic kidney disease (CKD) are now collectively called CKD-mineral bone disease (CKD-MBD). The pathophysiology of CKD-MBD is becoming clear with the emerging of αKlotho, originally identified as a progeria-causing protein, and bone-derived phosphaturic fibroblast growth factor 23 (FGF23) as associated factors. Meanwhile, compared with calcium and parathyroid hormone, which have long been linked with CKD-MBD, phosphate is now attracting more attention because of its association with complications and outcomes. Incidentally, as the pivotal roles of FGF23 and αKlotho in phosphate metabolism have been unveiled, how phosphate metabolism and hyperphosphatemia are involved in CKD-MBD and how they can be clinically treated have become of great interest. Thus, the aim of this review is reconsider CKD-MBD from the viewpoint of phosphorus, its involvement in the pathophysiology, causing complications, therapeutic approach based on the clinical evidence, and clarifying the importance of phosphorus management.
Collapse
Affiliation(s)
- Ken Tsuchiya
- Department of Blood Purification, Tokyo Women’s Medical University, Tokyo 162-8666, Japan
- Correspondence:
| | - Taro Akihisa
- Department of Nephrology, Tokyo Women’s Medical University, Tokyo 162-8666, Japan;
| |
Collapse
|
38
|
Xia J, Cao W. Epigenetic modifications of Klotho expression in kidney diseases. J Mol Med (Berl) 2021; 99:581-592. [PMID: 33547909 DOI: 10.1007/s00109-021-02044-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/10/2020] [Accepted: 01/20/2021] [Indexed: 12/21/2022]
Abstract
Developments of many renal diseases are substantially influenced by epigenetic modifications of numerous genes, mainly mediated by DNA methylations, histone modifications, and microRNA interference; however, not all gene modifications causally affect the disease onset or progression. Klotho is a critical gene whose repressions in various pathological conditions reportedly involve epigenetic regulatory mechanisms. Klotho is almost unexceptionally repressed early after acute or chronic renal injuries and its levels inversely correlated with the disease progression and severity. Moreover, the strategies of Klotho derepression via epigenetic modulations beneficially change the pathological courses both in vitro and in vivo. Hence, Klotho is not only considered a biomarker of the renal disease but also a potential or even an ideal target of therapeutic epigenetic intervention. Here, we summarize and discuss studies that investigate the Klotho repression and intervention in renal diseases from an epigenetic point of view. These information might shed new sights into the effective therapeutic strategies to prevent and treat various renal disorders.
Collapse
Affiliation(s)
- Jinkun Xia
- Center for Organ Fibrosis and Remodeling Research, Jiangsu Key Lab of Molecular Medicine, Nanjing University School of Medicine, Nanjing, China
| | - Wangsen Cao
- Center for Organ Fibrosis and Remodeling Research, Jiangsu Key Lab of Molecular Medicine, Nanjing University School of Medicine, Nanjing, China.
| |
Collapse
|
39
|
Mohanty SK, Suchiang K. Triiodothyronine (T3) enhances lifespan and protects against oxidative stress via activation of Klotho in Caenorhabditis elegans. Biogerontology 2021; 22:397-413. [PMID: 33851304 DOI: 10.1007/s10522-021-09923-0] [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: 01/23/2021] [Accepted: 04/07/2021] [Indexed: 10/21/2022]
Abstract
Age predisposes individuals to significant diseases, and the biological processes contributing to aging are currently under intense investigation. Klotho is an anti-aging protein with multifaceted roles and is an essential component of the endocrine fibroblast growth factor. In Caenorhabditis elegans (C. elegans), there are two prospective orthologs of α-Klotho, C50F7.10, and E02H9.5, identified. The two orthologs' products are homologous to the highly conserved KL1 domain of human and mouse Klotho protein. Considering the endocrine system's major involvement in an organism's homeostasis and that thyroid disorders increase with advancing age, the molecular mechanisms underlying its impact on different endocrine components during the aging process remain poorly characterized. In this study, we sought to determine the regulatory role of Triiodothyronine (T3) on homologs genes of klotho and its impact on different parameters of aging in the C. elegans model organism. We showed that T3 could increase the mRNA expressions of the klotho homologous genes in C. elegans. Moreover, T3 could also extend a worm lifespan and modulate oxidative stress resistance and aging biomarkers significantly and positively. Further investigations employing different mutant and transgenic strains reveal that these observed effects are mediated through the EGL-17/EGL-15 pathway via Klotho activation along with the involvement of transcription factor DAF-16. In conclusion, these findings have revealed an unexpected link between T3 and Klotho and how this link can modulate the aging process in C. elegans via activation of klotho. This study will help understand the crosstalk and regulations of different endocrine components and their consequences on the aging process in multiple species.
Collapse
Affiliation(s)
- Saswat Kumar Mohanty
- Department of Biochemistry and Molecular Biology, Pondicherry University, Pondicherry, 605 014, India
| | - Kitlangki Suchiang
- Department of Biochemistry and Molecular Biology, Pondicherry University, Pondicherry, 605 014, India.
| |
Collapse
|
40
|
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
|
41
|
Chen F, Gao Q, Wei A, Chen X, Shi Y, Wang H, Cao W. Histone deacetylase 3 aberration inhibits Klotho transcription and promotes renal fibrosis. Cell Death Differ 2021; 28:1001-1012. [PMID: 33024274 PMCID: PMC7937860 DOI: 10.1038/s41418-020-00631-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 02/05/2023] Open
Abstract
Development of renal fibrosis is a hallmark of renal aging and chronic kidney disease of all etiologies and characterized by extensive renal cell injuries and subsequent myofibroblast transdifferentiations (MTDs), which are significantly influenced by aberrant histone deacetylase (HDAC) activities. However, the key HDAC isoforms and effectors that are causally involved in the processes remain poorly understood. Here, we report that aberrant HDAC3 induction and its inhibition of Klotho, a renal epithelium-enriched aging suppressor, contribute significantly to renal fibrogenesis. HDAC3 was preferentially elevated with concomitant Klotho suppression in fibrotic kidneys incurred by unilateral ureter obstruction (UUO) and aristolochic acid nephropathy (AAN), whereas Hdac3 knockout resisted the fibrotic pathologies. The HDAC3 elevation is substantially blocked by the inhibitors of TGFβ receptor and Smad3 phosphorylation, suggesting that TGFβ/Smad signal activates Hdac3 transcription. Consistently, an HDAC3-selective inhibitor RGFP966 derepressed Klotho and mitigated the renal fibrotic injuries in both UUO and AAN mice. Further, HDAC3 overexpression or inhibition in renal epithelia inversely affected Klotho abundances and HDAC3 was inducibly associated with transcription regulators NCoR and NF-kB and bound to Klotho promoter in fibrotic kidney, supporting that aberrant HDAC3 targets and transcriptionally inhibits Klotho under renal fibrotic conditions. More importantly, the antirenal fibrosis effects of RGFP966 were largely compromised in mice with siRNA-mediated Klotho knockdown. Hence, HDAC3 aberration and the subsequent Klotho suppression constitute an important regulatory loop that promotes MTD and renal fibrosis and uses of HDAC3-selective inhibitors are potentially effective in treating renal fibrotic disorders.
Collapse
Affiliation(s)
- Fang Chen
- Center of Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University School of Medicine, Nanjing, China
| | - Qi Gao
- Center of Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University School of Medicine, Nanjing, China
| | - Ai Wei
- Center of Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University School of Medicine, Nanjing, China
| | - Xingren Chen
- Center of Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University School of Medicine, Nanjing, China
| | - Yujun Shi
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu, China
| | - Hongwei Wang
- Center of Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University School of Medicine, Nanjing, China.
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China.
| | - Wangsen Cao
- Center of Organ Fibrosis and Remodeling, Jiangsu Key Lab of Molecular Medicine, Nanjing University School of Medicine, Nanjing, China.
| |
Collapse
|
42
|
Lee J, Ju KD, Kim HJ, Tsogbadrakh B, Ryu H, Kang E, Kang M, Yang J, Kang HG, Ahn C, Oh KH. Soluble α-klotho anchors TRPV5 to the distal tubular cell membrane independent of FGFR1 by binding TRPV5 and galectin-1 simultaneously. Am J Physiol Renal Physiol 2021; 320:F559-F568. [PMID: 33615893 DOI: 10.1152/ajprenal.00044.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hypercalciuria is one of the early manifestations of diabetic nephropathy (DN). This is partially due to a decrease in the expression of renal transient receptor potential vanilloid type 5 (TRPV5), which is responsible for renal Ca2+ reabsorption. Soluble klotho has been previously determined to increase TRPV5 by cleaving sialic acid, causing TRPV5 to bind to membrane protein galectin-1. However, a recent study showed that soluble klotho binds to α2-3-sialyllactose, where sialic acid is located, on TRPV5, rather than cleave it. Here, we report that soluble klotho tethers TRPV5 on the membrane by binding both TRPV5 and galectin-1, thereby protecting membrane TRPV5 from diabetes-induced endocytosis. In the present study, we injected recombinant soluble α-klotho protein (rKL) into db/db and db/m mice for 8 wk and collected urine and kidneys. We administered rKL, AZD4547 [fibroblast growth factor (FGF) receptor type 1 inhibitor], and OTX008 (galectin-1 inhibitor) to cultured mouse distal tubular cells with or without 30 mM high-glucose (HG) exposure. db/db mice showed increased renal Ca2+ excretion and decreased renal TRPV5 expression. rKL treatment reversed this change. In vitro, TRPV5 expression in distal tubular cells decreased under HG conditions, and rKL successfully upregulated TRPV5 with or without FGF23. Also, immunofluorescence showed colocalization of klotho, TRPV5, and galectin-1 in distal tubule cells, suggesting that klotho binds to both TRPV5 and galectin-1. Moreover, when both FGF receptor type 1 and galectin-1 were inhibited, rKL failed to increase TRPV5 under HG conditions. Our results indicate that soluble klotho prevents TRPV5 from degradation and subsequent diabetes-induced endocytosis by anchoring TRPV5 through binding with both TRPV5 and galectin-1.NEW & NOTEWORTHY Soluble α-klotho anchors transient receptor potential vanilloid type 5 (TRPV5) on the apical membrane of the distal tubule by binding both TRPV5 and a membrane-abundant protein, galectin-1. This newly discovered mechanism works even when fibroblast growth factor (FGF)23 signaling is inhibited by treatment with FGF receptor type 1 inhibitor. Therefore, we identified how soluble α-klotho increases TRPV5 without FGF23. We confirmed this mechanism by observing that soluble α-klotho fails to enhance TRPV5 when both FGF receptor type 1 and galectin-1 are inhibited.
Collapse
Affiliation(s)
- Jinho Lee
- Center of Medical Innovation, Seoul National University Hospital, Seoul, Korea
| | - Kyung Don Ju
- Center of Medical Innovation, Seoul National University Hospital, Seoul, Korea
| | - Hyo Jin Kim
- Department of Internal Medicine, Pusan National University Hospital, Busan, Korea
| | | | - Hyunjin Ryu
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Eunjeong Kang
- Department of Internal Medicine, Ewha Womans University Seoul Hospital, Ewha Womans University College of Medicine, Seoul, South Korea
| | - Minjung Kang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jaeseok Yang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Transplantation Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Hee Gyung Kang
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea.,Department of Pediatrics, Seoul National University Children's Hospital, Seoul, Korea.,Kidney Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Curie Ahn
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Transplantation Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
43
|
Ewendt F, Feger M, Föller M. Role of Fibroblast Growth Factor 23 (FGF23) and αKlotho in Cancer. Front Cell Dev Biol 2021; 8:601006. [PMID: 33520985 PMCID: PMC7841205 DOI: 10.3389/fcell.2020.601006] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/15/2020] [Indexed: 12/16/2022] Open
Abstract
Together with fibroblast growth factors (FGFs) 19 and 21, FGF23 is an endocrine member of the family of FGFs. Mainly secreted by bone cells, FGF23 acts as a hormone on the kidney, stimulating phosphate excretion and suppressing formation of 1,25(OH)2D3, active vitamin D. These effects are dependent on transmembrane protein αKlotho, which enhances the binding affinity of FGF23 for FGF receptors (FGFR). Locally produced FGF23 in other tissues including liver or heart exerts further paracrine effects without involvement of αKlotho. Soluble Klotho (sKL) is an endocrine factor that is cleaved off of transmembrane Klotho or generated by alternative splicing and regulates membrane channels, transporters, and intracellular signaling including insulin growth factor 1 (IGF-1) and Wnt pathways, signaling cascades highly relevant for tumor progression. In mice, lack of FGF23 or αKlotho results in derangement of phosphate metabolism and a syndrome of rapid aging with abnormalities affecting most organs and a very short life span. Conversely, overexpression of anti-aging factor αKlotho results in a profound elongation of life span. Accumulating evidence suggests a major role of αKlotho as a tumor suppressor, at least in part by inhibiting IGF-1 and Wnt/β-catenin signaling. Hence, in many malignancies, higher αKlotho expression or activity is associated with a more favorable outcome. Moreover, also FGF23 and phosphate have been revealed to be factors relevant in cancer. FGF23 is particularly significant for those forms of cancer primarily affecting bone (e.g., multiple myeloma) or characterized by bone metastasis. This review summarizes the current knowledge of the significance of FGF23 and αKlotho for tumor cell signaling, biology, and clinically relevant parameters in different forms of cancer.
Collapse
Affiliation(s)
- Franz Ewendt
- Department of Nutritional Physiology, Institute of Agricultural and Nutritional Sciences, Martin-Luther University Halle-Wittenberg, Halle, Germany
| | - Martina Feger
- Department of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Michael Föller
- Department of Physiology, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
44
|
Dittmer KE, Heathcott RW, Marshall JC, Azarpeykan S. Expression of Phosphatonin-Related Genes in Sheep, Dog and Horse Kidneys Using Quantitative Reverse Transcriptase PCR. Animals (Basel) 2020; 10:ani10101806. [PMID: 33027890 PMCID: PMC7601102 DOI: 10.3390/ani10101806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/25/2020] [Accepted: 10/02/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Traditionally, it has been thought that control of body phosphorus was secondary to the tighter control of calcium. However, over the last 20 years, an extensive system for control of body phosphorus by proteins called phosphatonins has been shown to exist. Most research on phosphatonins has been done in rat or mouse models. This paper looks at whether important proteins and phosphorus channels in the phosphatonin pathways are present in the kidneys of dogs, horses and sheep. The results showed that all of the components of the phosphatonin system are present in these species, but that there are species differences in which protein or channel is most common, and in the relationships between the proteins and channels. This research is important because the phosphatonin system is involved in the progression of chronic kidney disease in humans and animals, and differences in the systems between animal species may affect treatment of chronic kidney disease. Abstract The aim of this preliminary study was to determine the relative expression of phosphatonin pathway-related genes in normal dog, sheep and horse kidneys and to explore the relationships between the different genes. Kidneys were collected post-mortem from 10 sheep, 10 horses and 8 dogs. RNA was extracted, followed by reverse transcriptase quantitative polymerase chain reaction for fibroblast growth factor receptor 1 IIIc (FGFR1IIIC), sodium-phosphate co-transporter (NPT) 1 (SLC17A1), NPT2a (SLC34A1), NPT2c (SLC34A3), parathyroid hormone 1 receptor (PTH1R), klotho (KL), vitamin D receptor (VDR), 1a-hydroxylase (CYP27B1) and 24-hydroxylase (CYP24A1). NPT2a was highly expressed in the dog kidneys, compared with those of the horses and sheep. NPT1 had greatest expression in horses and sheep, although the three different NPTs all had relatively similar expression in sheep. There was little variability in FGFR1IIIc expression, particularly in the dogs and horses. FGFR1IIIc expression was negatively correlated with NPT genes (except NPT2a in sheep), while NPT genes were all positively correlated with each other. Unexpectedly, klotho was positively correlated with NPT genes in all three species. These results provide the basis for further research into this important regulatory system. In particular, species differences in phosphatonin gene expression should be considered when considering the pathogenesis of chronic kidney disease.
Collapse
Affiliation(s)
- Keren E. Dittmer
- School of Veterinary Science, Massey University, Palmerston North 4410, New Zealand; (R.W.H.); (S.A.)
- Correspondence:
| | - Rosemary W. Heathcott
- School of Veterinary Science, Massey University, Palmerston North 4410, New Zealand; (R.W.H.); (S.A.)
| | - Jonathan C. Marshall
- School of Fundamental Sciences, Massey University, Palmerston North 4410, New Zealand;
| | - Sara Azarpeykan
- School of Veterinary Science, Massey University, Palmerston North 4410, New Zealand; (R.W.H.); (S.A.)
| |
Collapse
|
45
|
Xie T, Ye W, Liu J, Zhou L, Song Y. The Emerging Key Role of Klotho in the Hypothalamus-Pituitary-Ovarian Axis. Reprod Sci 2020; 28:322-331. [PMID: 32783104 DOI: 10.1007/s43032-020-00277-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/24/2020] [Indexed: 12/16/2022]
Abstract
The hypothalamus-pituitary-ovary axis is the most important system for regulating female reproductive endocrine function. Its dysfunction would lead to the abnormal secretion of gonadotropin-releasing hormone, follicle-stimulating hormone, or luteinizing hormone, and eventually result in the occurrence of reproductive disease, such as congenital hypogonadotropic hypogonadism, polycystic ovary syndrome, and premature ovarian failure. Recently, an anti-aging gene, Klotho, has gained broad attention in female reproductive diseases. Reports have shown that Klotho is closely correlated to the hypothalamus-pituitary-ovary axis and plays a key role in the development and progression of reproductive diseases. With this issue, we generally review the physiological and pathological role of Klotho in the hypothalamus-pituitary-ovary axis. We also review the underlying mechanisms of Klotho in promoting and preventing female reproductive diseases, which involve the dysfunction of the fibroblast growth factor-Klotho endocrine system, the abnormal signaling regulation of Wnt-β-catenin and insulin-like growth factor-1, the accumulation of oxidative stress, and the inhibition of autophagy, eventually affecting the genesis, development, ovulation, or atresia of follicles. The present review would provide new insights and potential therapeutic target strategies for clinical strategies.
Collapse
Affiliation(s)
- Tingting Xie
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China
| | - Wenting Ye
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China
| | - Jing Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China.
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
| | - Yali Song
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China.
| |
Collapse
|
46
|
Klotho rewires cellular metabolism of breast cancer cells through alteration of calcium shuttling and mitochondrial activity. Oncogene 2020; 39:4636-4649. [PMID: 32398866 DOI: 10.1038/s41388-020-1313-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 02/07/2023]
Abstract
Klotho is a transmembrane protein, which can be shed and act as a circulating hormone and is involved in regulating cellular calcium levels and inhibition of the PI3K/AKT pathway. As a longevity hormone, it protects normal cells from oxidative stress, and as a tumor suppressor it inhibits growth of cancer cells. Mechanisms governing these differential activities have not been addressed. Altered cellular metabolism is a hallmark of cancer and dysregulation of mitochondrial activity is a hallmark of aging. We hypothesized that klotho exerts its differential effects through regulation of these two hallmarks. Treatment with klotho inhibited glycolysis, reduced mitochondrial activity and membrane potential only in cancer cells. Accordingly, global metabolic screen revealed that klotho altered pivotal metabolic pathways, amongst them glycolysis and tricarboxylic acid cycle in breast cancer cells. Alteration of metabolic activity and increased AMP/ATP ratio lead to LKB1-dependent AMPK activation. Indeed, klotho induced AMPK phosphorylation; furthermore, inhibition of LKB1 partially abolished klotho's tumor suppressor activity. By diminishing deltapsi (Δψ) klotho also inhibited mitochondria Ca2+ shuttling thereby impairing mitochondria communication with SOCE leading to reduced Ca2+ influx by SOCE channels. The reduced SOCE was followed by ER Ca2+ depletion and stress. These data delineate mechanisms mediating the differential effects of klotho toward cancer versus normal cells, and indicate klotho as a potent regulator of metabolic activity.
Collapse
|
47
|
Shardell M, Drew DA, Semba RD, Harris TB, Cawthon PM, Simonsick EM, Kalyani RR, Schwartz AV, Kritchevsky SB, Newman AB. Plasma Soluble αKlotho, Serum Fibroblast Growth Factor 23, and Mobility Disability in Community-Dwelling Older Adults. J Endocr Soc 2020; 4:bvz032. [PMID: 32405607 PMCID: PMC7209777 DOI: 10.1210/jendso/bvz032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 04/20/2020] [Indexed: 12/13/2022] Open
Abstract
CONTEXT αKlotho is a hormone and co-receptor for fibroblast growth factor 23 (FGF23), a hormone that downregulates active vitamin D synthesis and promotes phosphate excretion. Low αKlotho and high FGF23 occur in chronic kidney disease (CKD). OBJECTIVE We aimed to assess the relationships of αKlotho and FGF23 with mobility disability in community-dwelling older adults. DESIGN AND SETTING We estimated associations of plasma-soluble αKlotho and serum FGF23 concentrations with mobility disability over 6 years. Additional analyses was stratified by CKD. PARTICIPANTS Participants included 2751 adults (25.0% with CKD), aged 71 to 80 years, from the 1998 to 1999 Health, Aging, and Body Composition Study visit. MAIN OUTCOME MEASURES Walking disability and stair climb disability were defined as self-reported "a lot of difficulty" or an inability to walk a quarter mile and climb 10 stairs, respectively. RESULTS Median (interquartile range [IQR]) serum FGF23 and plasma soluble αKlotho concentrations were 46.6 (36.7, 60.2) pg/mL and 630.4 (478.4, 816.0) pg/mL, respectively. After adjustment, higher αKlotho concentrations were associated with lower walking disability rates (Rate Ratio [RR] highest vs. lowest tertile = 0.74; 95% confidence interval l [CI] = 0.62, 0.89; P = 0.003). Higher FGF23 concentrations were associated with higher walking disability rates (RR highest vs. lowest tertile = 1.24; 95%CI = 1.03, 1.50; P = 0.005). Overall, higher αKlotho combined with lower FGF23 was associated with the lowest walking disability rates (P for interaction = 0.023). Stair climb disability findings were inconsistent. No interactions with CKD were statistically significant (P for interaction > 0.10). CONCLUSIONS Higher plasma soluble αKlotho and lower serum FGF23 concentrations were associated with lower walking disability rates in community-dwelling older adults, particularly those without CKD.
Collapse
Affiliation(s)
- Michelle Shardell
- Institute for Genome Sciences, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland
| | - David A Drew
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Richard D Semba
- Department of Ophthalmology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Science, National Institute on Aging Intramural Research Program, Bethesda, Maryland
| | - Peggy M Cawthon
- Research Institute, California Pacific Medical Center, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California
| | - Eleanor M Simonsick
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, Baltimore, Maryland
| | - Rita R Kalyani
- Department of Endocrinology, Metabolism, and Diabetes, Johns Hopkins Medical Institutions, Baltimore Maryland
| | - Ann V Schwartz
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California
| | - Stephen B Kritchevsky
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest University, Winston-Salem, North Carolina
| | - Anne B Newman
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
48
|
Imenez Silva PH, Katamesh-Benabbas C, Chan K, Pastor Arroyo EM, Knöpfel T, Bettoni C, Ludwig MG, Gasser JA, Brandao-Burch A, Arnett TR, Bonny O, Seuwen K, Wagner CA. The proton-activated ovarian cancer G protein-coupled receptor 1 (OGR1) is responsible for renal calcium loss during acidosis. Kidney Int 2020; 97:920-933. [DOI: 10.1016/j.kint.2019.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 11/29/2019] [Accepted: 12/05/2019] [Indexed: 12/19/2022]
|
49
|
Dhayat NA, Pruijm M, Ponte B, Ackermann D, Leichtle AB, Devuyst O, Ehret G, Guessous I, Pechère-Bertschi A, Pastor J, Martin PY, Burnier M, Fiedler GM, Vogt B, Moe OW, Bochud M, Fuster DG. Parathyroid Hormone and Plasma Phosphate Are Predictors of Soluble α-Klotho Levels in Adults of European Descent. J Clin Endocrinol Metab 2020; 105:5644279. [PMID: 31774122 PMCID: PMC7341480 DOI: 10.1210/clinem/dgz232] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 11/26/2019] [Indexed: 01/08/2023]
Abstract
CONTEXT α-klotho is an integral membrane protein that serves as a coreceptor for fibroblast growth factor 23 (FGF23) in conjunction with cognate fibroblast growth factor receptors. Proteolytic cleavage sheds the ectodomain of α-klotho (soluble α-klotho) as an endocrine substance into blood, urine, and cerebrospinal fluid. OBJECTIVE To study the relationship of soluble α-klotho to mineral metabolism in the general population with mainly preserved kidney function. DESIGN Cross-sectional analysis of the associations between soluble α-klotho with laboratory markers of markers of mineral metabolism in a population-based cohort. SETTING Three centers in Switzerland including 1128 participants. MEASURES Soluble full-length α-klotho levels by a specific immunoassay and markers of mineral metabolism. RESULTS The median serum level of soluble α-klotho was 15.0 pmol/L. Multivariable analyses using α-klotho as the outcome variable revealed a sex-by-PTH interaction: In men, PTH was positively associated with α-klotho levels, whereas this association was negative in women. Plasma phosphate associated with soluble α-klotho levels in an age-dependent manner, changing from a positive association in young adults gradually to a negative association in the elderly. The decline of 1,25 (OH)2 vitamin D3 levels in parallel to the gradual impairment of kidney function was greatly attenuated in the setting of high circulating soluble α-klotho levels. CONCLUSIONS Soluble α-klotho level is associated with plasma phosphate in an age-dependent manner and with PTH in a sex-dependent manner. Furthermore, our data reveal soluble α-klotho as a modulator of 1,25 (OH)2 vitamin D3 levels in individuals with preserved renal function.
Collapse
Affiliation(s)
- Nasser A Dhayat
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Menno Pruijm
- Nephrology Service, Lausanne University Hospital, Lausanne, Switzerland
| | - Belen Ponte
- Nephrology Service, Department of Specialties of Internal Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Daniel Ackermann
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Alexander Benedikt Leichtle
- Department of Laboratory Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Insel Data Science Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Georg Ehret
- Cardiology Service, Department of Specialties of Internal Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Idris Guessous
- Division of Primary Care Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Antoinette Pechère-Bertschi
- Endocrinology Service, Department of Specialties of Internal Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Johanne Pastor
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, Department of Physiology and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Pierre-Yves Martin
- Nephrology Service, Department of Specialties of Internal Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Michel Burnier
- Nephrology Service, Lausanne University Hospital, Lausanne, Switzerland
| | - Georg-Martin Fiedler
- Department of Laboratory Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Bruno Vogt
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Orson W Moe
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, Department of Physiology and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Murielle Bochud
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Daniel G Fuster
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Correspondence and Reprint Requests: Daniel G. Fuster, Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 15, 3010 Bern, Switzerland. E-mail:
| |
Collapse
|
50
|
Zhong X, Jagarlapudi S, Weng Y, Ly M, Rouse JC, McClure K, Ishino T, Zhang Y, Sousa E, Cohen J, Tzvetkova B, Cote K, Scarcelli JJ, Johnson K, Palandra J, Apgar JR, Yaddanapudi S, Gonzalez-Villalobos RA, Opsahl AC, Lam K, Yao Q, Duan W, Sievers A, Zhou J, Ferguson D, D'Antona A, Zollner R, Zhu HL, Kriz R, Lin L, Clerin V. Structure-function relationships of the soluble form of the antiaging protein Klotho have therapeutic implications for managing kidney disease. J Biol Chem 2020; 295:3115-3133. [PMID: 32005658 PMCID: PMC7062171 DOI: 10.1074/jbc.ra119.012144] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/20/2020] [Indexed: 01/28/2023] Open
Abstract
The fortuitously discovered antiaging membrane protein αKlotho (Klotho) is highly expressed in the kidney, and deletion of the Klotho gene in mice causes a phenotype strikingly similar to that of chronic kidney disease (CKD). Klotho functions as a co-receptor for fibroblast growth factor 23 (FGF23) signaling, whereas its shed extracellular domain, soluble Klotho (sKlotho), carrying glycosidase activity, is a humoral factor that regulates renal health. Low sKlotho in CKD is associated with disease progression, and sKlotho supplementation has emerged as a potential therapeutic strategy for managing CKD. Here, we explored the structure-function relationship and post-translational modifications of sKlotho variants to guide the future design of sKlotho-based therapeutics. Chinese hamster ovary (CHO)- and human embryonic kidney (HEK)-derived WT sKlotho proteins had varied activities in FGF23 co-receptor and β-glucuronidase assays in vitro and distinct properties in vivo Sialidase treatment of heavily sialylated CHO-sKlotho increased its co-receptor activity 3-fold, yet it remained less active than hyposialylated HEK-sKlotho. MS and glycopeptide-mapping analyses revealed that HEK-sKlotho is uniquely modified with an unusual N-glycan structure consisting of N,N'-di-N-acetyllactose diamine at multiple N-linked sites, one of which at Asn-126 was adjacent to a putative GalNAc transfer motif. Site-directed mutagenesis and structural modeling analyses directly implicated N-glycans in Klotho's protein folding and function. Moreover, the introduction of two catalytic glutamate residues conserved across glycosidases into sKlotho enhanced its glucuronidase activity but decreased its FGF23 co-receptor activity, suggesting that these two functions might be structurally divergent. These findings open up opportunities for rational engineering of pharmacologically enhanced sKlotho therapeutics for managing kidney disease.
Collapse
Affiliation(s)
- Xiaotian Zhong
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139.
| | - Srinath Jagarlapudi
- Internal Medicine, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Yan Weng
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Mellisa Ly
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts 01810
| | - Jason C Rouse
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts 01810
| | - Kim McClure
- Internal Medicine, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Tetsuya Ishino
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Yan Zhang
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Eric Sousa
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Justin Cohen
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Boriana Tzvetkova
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts 01810
| | - Kaffa Cote
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts 01810
| | - John J Scarcelli
- Cell Line Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts 01810
| | - Keith Johnson
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts 01810
| | - Joe Palandra
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - James R Apgar
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Suma Yaddanapudi
- Internal Medicine, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | | | - Alan C Opsahl
- Internal Medicine, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Khetemenee Lam
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Qing Yao
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Weili Duan
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Annette Sievers
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Jing Zhou
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Darren Ferguson
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Aaron D'Antona
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Richard Zollner
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Hongli L Zhu
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Ron Kriz
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Laura Lin
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Valerie Clerin
- Internal Medicine, Pfizer Worldwide Research, Cambridge, Massachusetts 02139.
| |
Collapse
|