51
|
Montoro-Molina S, López-Carmona A, Quesada A, O'Valle F, Martín-Morales N, Osuna A, Vargas F, Wangensteen R. Klotho and Aminopeptidases as Early Biomarkers of Renal Injury in Zucker Obese Rats. Front Physiol 2018; 9:1599. [PMID: 30483154 PMCID: PMC6243116 DOI: 10.3389/fphys.2018.01599] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/24/2018] [Indexed: 01/08/2023] Open
Abstract
The aim of this study was to investigate if urinary glutamyl aminopeptidase (GluAp), alanyl aminopeptidase (AlaAp), Klotho and hydroxyproline can be considered as potential biomarkers of renal injury and fibrosis in an experimental model of obesity. Male Zucker lean (ZL) and obese (ZO) rats were studied from 2 to 8 months old. Kidneys from ZO rats at the end of the study (8 months old) developed mild focal and segmental glomerulosclerosis as well as moderate tubulointerstitial injury. Urinary excretion of Klotho was higher in ZO rats at 2, 5, and 8 months of study, plasma Klotho levels were reduced and protein abundance of Klotho in renal tissue was similar in ZL and ZO rats. GluAp and AlaAp urinary activities were also increased in ZO rats throughout the time-course study. ZO rats showed an augmentation of hydroxyproline content in renal tissue and a significant increase of tubulointerstitial fibrosis. Correlation studies demonstrated that GluAp, AlaAp, and Klotho are early diagnostic markers of renal lesions in Zucker obese rats. Proteinuria and hydroxyproline can be considered delayed diagnostic markers because their contribution to diagnosis starts later. Another relevant result is that GluAp, AlaAp, and Klotho are related not only with diagnosis but also with prognosis of renal lesions in Zucker obese rats. Moreover, strong predictive correlations of aminopeptidasic activities with the percentage of renal fibrosis or with renal hydroxyproline content at the end of the experiment were observed, indicating that an early increased excretion of these markers is related with a higher later extent of fibrosis in Zucker obese rats. In conclusion, GluAp, AlaAp, and Klotho are early diagnostic markers that are also related with the extent of renal fibrosis in Zucker obese rats. Therefore, they have a potential use not only in diagnosis, but also in prognosis of obesity-associated renal lesions.
Collapse
Affiliation(s)
| | | | - Andrés Quesada
- Servicio de Nefrología, Unidad Experimental, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Francisco O'Valle
- Departamento de Anatomía Patológica, Instituto de Biopatología y Medicina Regenerativa (IBIMER), Facultad de Medicina, Universidad de Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Natividad Martín-Morales
- Departamento de Anatomía Patológica, Instituto de Biopatología y Medicina Regenerativa (IBIMER), Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Antonio Osuna
- Servicio de Nefrología, Unidad Experimental, Hospital Universitario Virgen de las Nieves, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Félix Vargas
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | | |
Collapse
|
52
|
Abstract
Chronic kidney disease (CKD) is an inherently systemic disease that refers to a long-term loss of kidney function. The progression of CKD has repercussions for other organs, leading to many kinds of extrarenal complications. Intensive studies are now being undertaken to reveal the risk factors and pathophysiological mechanism of this disease. During the past 20 years, increasing evidence from clinical and basic studies has indicated that klotho, which was initially known as an anti-aging gene and is mainly expressed in the kidney, is significantly correlated with the development and progression of CKD and its complications. Here, we discuss in detail the role and pathophysiological implications of klotho in ion disorders, the inflammation response, vascular calcification, mineral bone disorders, and renal fibrosis in CKD. Based on the pathogenic mechanism of klotho deficiency and klotho decline in urine early in CKD stage 2 and even earlier in CKD stage 1, it is not difficult to understand that soluble klotho can serve as an early and sensitive marker of CKD. Moreover, the prevention of klotho decline by several mechanisms can attenuate renal injuries, retard CKD progression, ameliorate extrarenal complications, and improve renal function. In this review, we focus on the functions and pathophysiological implications of klotho in CKD and its extrarenal complications as well as its potential applications as a diagnostic and/or prognostic biomarker for CKD and as a novel treatment strategy to improve and decrease the burden of comorbidity in CKD.
Collapse
|
53
|
Zhang L, Liu T. Clinical implication of alterations in serum Klotho levels in patients with type 2 diabetes mellitus and its associated complications. J Diabetes Complications 2018; 32:922-930. [PMID: 30042059 DOI: 10.1016/j.jdiacomp.2018.06.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 06/05/2018] [Accepted: 06/06/2018] [Indexed: 02/05/2023]
Abstract
AIM To investigate the clinical significance of serum α-Klotho and β-Klotho levels in patients with type 2 diabetes mellitus (T2DM) and its associated complications. METHODS Serum α-Klotho and β-Klotho levels were measured using an ELISA kit in 817 individuals, including 127 with T2DM, 106 with diabetic nephropathy, 99 with diabetic retinopathy, 108 with diabetic neuropathy, 102 with diabetic foot disease, 135 with T2DM and more than one complication and 140 healthy controls. RESULTS Both α-Klotho and β-Klotho levels were significantly decreased in the T2DM group and the groups with associated complications compared with the levels in control group. The differences between the T2DM group and the T2DM with complications groups were not significant, except between the diabetic nephropathy group and the other diabetic complications groups. In addition, α-Klotho and β-Klotho levels were negatively correlated with serum fructosamine and HbA1c but were not associated with serum glucose in the model including all participants. Moreover, decreases in α-Klotho and β-Klotho levels in the high glucose-exposed cell culture model, which was dependent on glucose exposure time, were confirmed. CONCLUSIONS Levels of α-Klotho and β-Klotho were downregulated in patients in the T2DM and complications groups. Our findings indicate that serum Klotho levels were associated with the development of T2DM, and long-term control of blood glucose will be beneficial in ameliorating changes to α-Klotho and β-Klotho levels in patients with T2DM and complications.
Collapse
Affiliation(s)
- Lingling Zhang
- Department of Laboratory, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, PR China
| | - Tianming Liu
- Department of Laboratory, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, PR China.
| |
Collapse
|
54
|
Behringer V, Stevens JMG, Deschner T, Sonnweber R, Hohmann G. Aging and sex affect soluble alpha klotho levels in bonobos and chimpanzees. Front Zool 2018; 15:35. [PMID: 30250491 PMCID: PMC6146871 DOI: 10.1186/s12983-018-0282-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/07/2018] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Throughout life, physiological homeostasis is challenged and the capacity to cope with such challenges declines with increasing age. In many species, sex differences exist in life expectancy. Sex-specific differences have been related to extrinsic factors like mate competition and/or intrinsic proximate mechanisms such as hormonal changes. In humans, an intrinsic factor related to aging is soluble alpha klotho (α-Kl). Both sexes show an age-related decline in α-Kl, but throughout life women have higher levels than men of the same age. Sex differences in α-Kl have been linked to a shorter lifespan, as well as to specific morbidity factors such as atherosclerosis and arteries calcifications. In non-human animals, information on α-Kl levels is rare and restricted to experimental work. Our cross-sectional study is the first on α-Kl levels in two long-lived species: bonobos (Pan paniscus) and chimpanzees (Pan troglodytes). As in most mammals, female bonobos and chimpanzees have longer life expectancy than males. METHODS We measured serum α-Kl levels of 140 subjects from 16 zoos with an ELISA to examine if α-Kl levels reflect this difference in life expectancy. RESULTS In both species and in both sexes, α-Kl levels declined with age suggesting that this marker has potential for aging studies beyond humans. We also found species-specific differences. Adult female bonobos had higher α-Kl levels than males, a difference that corresponds to the pattern found in humans. In chimpanzees, we found the opposite: males had higher α-Kl levels than females. CONCLUSION We suggest that contrasting sex differences in adult α-Kl levels mirror the dominance relations between females and males of the two Pan species; and that this might be related to corresponding sex differences in their exposure to stress. In humans, higher cortisol levels were found to be related to lower α-Kl levels. We conclude that there is great potential for studying aging processes in hominoids, and perhaps also in other non-human primates, by measuring α-Kl levels. To better understand the causes for sex differences in this aging marker, consideration of behavioural parameters such as competition and stress exposure will be required as well as other physiological markers.
Collapse
Affiliation(s)
- V. Behringer
- Department of Primatology, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - J. M. G. Stevens
- Antwerp Zoo Centre for Research and Conservation, Royal Zoological Society of Antwerp, K. Astridplein 26, 2018 Antwerp, Belgium
- Behavioral Ecology and Ecophysiology, Department of Biology, University of Antwerp, 2610 Wilrijk, Belgium
| | - T. Deschner
- Department of Primatology, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - R. Sonnweber
- Department of Primatology, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - G. Hohmann
- Department of Primatology, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| |
Collapse
|
55
|
Correlation between Soluble α-Klotho and Renal Function in Patients with Chronic Kidney Disease: A Review and Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9481475. [PMID: 30159331 PMCID: PMC6109492 DOI: 10.1155/2018/9481475] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/29/2018] [Accepted: 07/11/2018] [Indexed: 01/11/2023]
Abstract
Objective Over decades, numerous inconsistent studies are reported on the relationship between soluble α-Klotho and renal function in patients with chronic kidney disease (CKD). This study aims to perform a meta-analysis to figure out the correlations between soluble α-Klotho and renal function in patients with CKD. Materials and Methods We searched medical and scientific literature databases, PubMed and EMBASE (from the inception to October 2017), for publications that reported studies on associations between soluble α-Klotho and renal function in patients with CKD. Only publications in English were extracted. Summary correlation coefficient (r) values were extracted from each study, and 95% confidence intervals (CIs) were calculated. Publication bias was tested, and sensitivity and subgroup analyses were performed to investigate potential heterogeneity. Results Of 611 studies, 9 publications with 1457 patients were included into the analysis. The following data were extracted from the literature: first author, year of publication, research region, research index, sample size, average age and Pearson or Spearman correlation coefficient, study design, the αKlotho/FGF23 assays utilized, full length, or the C-terminal fragment of FGF23. The pooled r between α-Klotho and estimated glomerular filtration rate (eGFR), FGF-23 were 0.35 (95%CI, 0.23~0.46, and P<0.05), -0.10 (95%CI, -0.19~-0.01, and P<0.05) with remarkable significance, indicating moderate heterogeneity. There was no significant heterogeneity between subgroups in analyses of α-Klotho and eGFR stratified by research region, mean age, and eGFR, but heterogeneity exists in analyses of α-Klotho and FGF-23 stratified by research region. There was no significant correlation between a-klotho and Ca and PTH and PHOS. There was no evidence of publication bias with Egger's test (p=0.360) or with Begg's test (p=0.902) and the distribution of funnel plots was symmetrical in all of our analysis. Conclusions There exists a significant positive correlation between soluble α-Klotho and eGFR in patients with CKD. Also, a significant negative correlation between α-Klotho and FGF23 levels is proven. This raises hope to employ αKlotho and FGF23 as early biomarkers of CKD. However, further large prospective follow-up researches are needed to validate this hypothesis and to explore whether maintaining or elevating the Klotho level could improve renal function and complications in CKD patients.
Collapse
|
56
|
Kuriyama N, Ozaki E, Mizuno T, Ihara M, Mizuno S, Koyama T, Matsui D, Watanabe I, Akazawa K, Takeda K, Takada A, Inaba M, Yamada S, Motoyama K, Takeshita W, Iwai K, Hashiguchi K, Kobayashi D, Kondo M, Tamura A, Yamada K, Nakagawa M, Watanabe Y. Association between α-Klotho and Deep White Matter Lesions in the Brain: A Pilot Case Control Study Using Brain MRI. J Alzheimers Dis 2018; 61:145-155. [PMID: 29154273 DOI: 10.3233/jad-170466] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND The anti-aging protein, α-Klotho, may be involved in cognitive decline and has potential as a surrogate marker that reflects dementia. However, the role of α-Klotho in the brain has not been sufficiently investigated. OBJECTIVE Here, we investigated the association between α-Klotho and cognitive decline that is associated with cerebral deep white matter lesions (DWMLs). METHODS Two hundred-eighty participants (187 males and 93 females, mean age: 70.8 years old) were evaluated for DWMLs, and the Fazekas scale (Grade) was assessed following brain magnetic resonance imaging. A questionnaire concerning lifestyle and neuropsychological tests was administered, and their associations with the blood α-Klotho level were retrospectively investigated. RESULTS The α-Klotho level was 685.1 pg/mL in Grade 0 (68 subjects), 634.1 in G1 (134), 596.0 in G2 (62), and 571.6 in G3 (16), showing that the level significantly decreased with advanced grades. Significant correlations were noted between the α-Klotho level and higher brain function tests including the Mini-Mental State Examination and word fluency tests (p < 0.05). When a 90th percentile value of the level in the G0 group (400 pg/mL) or lower was defined as a low α-Klotho level, the odds ratio of the high-grade G3 group was 2.9 (95% confidence interval: 1.4-7.8) (after correction for age, sex, hypertension, and chronic kidney disease), which was significant. CONCLUSION A reduced blood α-Klotho level was correlated with grading of cerebral DWMLs and was accompanied by cognitive decline as an independent risk factor. The α-Klotho level may serve as a useful clinical index of vascular cognitive impairment.
Collapse
Affiliation(s)
- Nagato Kuriyama
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Etsuko Ozaki
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshiki Mizuno
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masafumi Ihara
- Division of Neurology, Department of Stroke and Cerebrovascular Diseases, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Shigeto Mizuno
- Department of Endoscopy, Kindai University Nara Hospital, Japan
| | - Teruhide Koyama
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Matsui
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Isao Watanabe
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kentaro Akazawa
- Department of Radiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuo Takeda
- Kyoto Industrial Health Association, Kyoto, Japan
| | | | - Masaaki Inaba
- Department of Metabolism, Endocrinology, and Molecular Medicine, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Shinsuke Yamada
- Department of Metabolism, Endocrinology, and Molecular Medicine, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Koka Motoyama
- Department of Metabolism, Endocrinology, and Molecular Medicine, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Wakiko Takeshita
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Komei Iwai
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kanae Hashiguchi
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daiki Kobayashi
- Department of Medicine, Division of General Internal Medicine, St. Luke's International Hospital, Tokyo, Japan
| | - Masaki Kondo
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Aiko Tamura
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kei Yamada
- Department of Radiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masanori Nakagawa
- North Medical Center, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshiyuki Watanabe
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
57
|
Kimura T, Shiizaki K, Akimoto T, Shinzato T, Shimizu T, Kurosawa A, Kubo T, Nanmoku K, Kuro-O M, Yagisawa T. The impact of preserved Klotho gene expression on antioxidative stress activity in healthy kidney. Am J Physiol Renal Physiol 2018; 315:F345-F352. [PMID: 29693450 DOI: 10.1152/ajprenal.00486.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Klotho, which was originally identified as an antiaging gene, forms a complex with fibroblast growth factor 23 receptor in the kidney, with subsequent signaling that regulates mineral metabolism. Other biological activities of Klotho, including antiaging effects such as protection from various types of cellular stress, have been shown; however, the precise mechanism of these effects of Klotho gene in the healthy human kidney is not well understood. In this study, we examined the relationships of Klotho and antioxidative stress gene expression levels in zero-hour biopsy specimens from 44 donors in kidney transplantation and verified them in animal models whose Klotho gene expression levels were varied. The nitrotyrosine expression level in the kidney was evaluated in these animal models. Expression levels of Klotho gene were positively correlated with the p53 gene and antioxidant enzyme genes such as catalase, superoxide dismutase 1 (SOD1), SOD2, peroxiredoxin 3 (PRDX3), and glutathione peroxidase 1 (GPX1) but not clinical parameters such as age and renal function or pathological features such as glomerulosclerosis and interstitial fibrosis tubular atrophy. The expression levels of all genes were significantly higher in mice with Klotho overexpression than in wild-type mice, and those except for catalase, PRDX3, and GPX1 were significantly lower in Klotho-deficient mice than in wild-type littermate mice. Nitrotyrosine-positive bands of various sizes were observed in kidney from Klotho-deficient mice only. The preservation of Klotho gene expression might induce the antioxidative stress mechanism for homeostasis of healthy human kidney independently of its general condition, including age, renal function, and histological findings.
Collapse
Affiliation(s)
- Takaaki Kimura
- Division of Renal Surgery and Transplantation, Department of Urology, Jichi Medical University, Shimotsuke, Tochigi , Japan
| | - Kazuhiro Shiizaki
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi , Japan
| | - Tetsu Akimoto
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi , Japan
| | - Takahiro Shinzato
- Division of Renal Surgery and Transplantation, Department of Urology, Jichi Medical University, Shimotsuke, Tochigi , Japan
| | - Toshihiro Shimizu
- Division of Renal Surgery and Transplantation, Department of Urology, Jichi Medical University, Shimotsuke, Tochigi , Japan
| | - Akira Kurosawa
- Division of Renal Surgery and Transplantation, Department of Urology, Jichi Medical University, Shimotsuke, Tochigi , Japan
| | - Taro Kubo
- Division of Renal Surgery and Transplantation, Department of Urology, Jichi Medical University, Shimotsuke, Tochigi , Japan
| | - Koji Nanmoku
- Division of Renal Surgery and Transplantation, Department of Urology, Jichi Medical University, Shimotsuke, Tochigi , Japan
| | - Makoto Kuro-O
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi , Japan
| | - Takashi Yagisawa
- Division of Renal Surgery and Transplantation, Department of Urology, Jichi Medical University, Shimotsuke, Tochigi , Japan
| |
Collapse
|
58
|
Cho NJ, Han DJ, Lee JH, Jang SH, Kang JS, Gil HW, Park S, Lee EY. Soluble klotho as a marker of renal fibrosis and podocyte injuries in human kidneys. PLoS One 2018; 13:e0194617. [PMID: 29590173 PMCID: PMC5874023 DOI: 10.1371/journal.pone.0194617] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/06/2018] [Indexed: 11/26/2022] Open
Abstract
Klotho deficiency is relevant to renal fibrosis and podocyte injury in vivo and in vitro. We examined whether histological findings of renal biopsy specimens were associated with the levels of soluble klotho in humans. We investigated renal biopsy specimens of 67 patients and detailed microscopic findings were reviewed. Soluble serum/urinary klotho and urinary angiotensinogen were assessed by enzyme-linked immunosorbent assays, and tissue klotho expression was assessed by immunohistochemical staining. The median age of the study participants was 35.6 years. High serum klotho levels (≥14 pg/mL) were associated with decreased odds ratios (ORs) of interstitial fibrosis (OR = 0.019, P = 0.003) and segmental sclerosis (OR = 0.190, P = 0.022) in multivariable logistic regression analysis. Patients with a lower urinary klotho-to-creatinine ratio (UKCR) were significantly more likely to have diffuse foot process effacement (OR = 0.450, P = 0.010). The area under the receiver-operating characteristic curve (AUC) of serum klotho for predicting interstitial fibrosis was 0.920 (95% CI, 0.844–0.996), and the best cut-off value of serum klotho was 138.1 pg/mL. The AUC of UKCR for predicting diffuse foot process effacement was 0.754 (95% CI, 0.636–0.872), and the best cut-off value of UKCR was 96.7 pg/mgCr. Urinary angiotensinogen-to-creatinine ratio was not associated with serum klotho, UKCR, or any pathological finding. Our data suggested that soluble serum and urinary klotho levels represent a potential biomarker to predict renal fibrosis and podocyte injury in humans.
Collapse
Affiliation(s)
- Nam-Jun Cho
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Dong-Jae Han
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Ji-Hye Lee
- Department of Pathology, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Si-Hyong Jang
- Department of Pathology, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Jeong Suk Kang
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Hyo-Wook Gil
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Samel Park
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Eun Young Lee
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
- * E-mail:
| |
Collapse
|
59
|
Rowland J, Akbarov A, Maan A, Eales J, Dormer J, Tomaszewski M. Tick-Tock Chimes the Kidney Clock - from Biology of Renal Ageing to Clinical Applications. Kidney Blood Press Res 2018; 43:55-67. [PMID: 29402805 DOI: 10.1159/000486907] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 01/18/2018] [Indexed: 11/19/2022] Open
Abstract
Ageing of the kidney is a multi-dimensional process that occurs simultaneously at the molecular, cellular, histological, anatomical and physiological level. Nephron number and renal cortical volume decline, renal tubules become atrophic and glomeruli become sclerotic with age. These structural changes are accompanied by a decline in glomerular filtration rate, decreased sodium reabsorption and potassium excretion, reduced urinary concentrating capacity and alterations in the endocrine activity of the kidney. However, the pace of progression of these changes is not identical in everyone - individuals of the same age and seemingly similar clinical profile often exhibit stark differences in the age-related decline in renal health. Thus, chronological age poorly reflects the time-dependent changes that occur in the kidney. An ideal measure of renal vitality is biological kidney age - a measure of the age-related changes in physiological function. Replacing chronological age with biological age could provide numerous clinical benefits including improved prognostic accuracy in renal transplantation, better stratification of risk and identification of those who are on a fast trajectory to an age-related drop in kidney health.
Collapse
Affiliation(s)
- Joshua Rowland
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, United Kingdom
| | - Artur Akbarov
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, United Kingdom
| | - Akhlaq Maan
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, United Kingdom
| | - James Eales
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, United Kingdom
| | - John Dormer
- University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, United Kingdom.,Division of Medicine, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
60
|
Tsai KD, Lee WX, Chen W, Chen BY, Chen KL, Hsiao TC, Wang SH, Lee YJ, Liang SY, Shieh JC, Lin TH. Upregulation of PRMT6 by LPS suppresses Klotho expression through interaction with NF-κB in glomerular mesangial cells. J Cell Biochem 2018; 119:3404-3416. [PMID: 29131380 DOI: 10.1002/jcb.26511] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 11/09/2017] [Indexed: 01/13/2023]
Abstract
Lipopolysaccharide (LPS) released from gram-negative bacteria stimulates immune responses in infected cells. Epigenetic modifications such as DNA methylation and protein methylation modulate LPS-induced innate immune gene expressions. Expression of the Klotho protein decreased with LPS treatment in rats. In a cellular model, information regarding the effect of LPS on Klotho expression was meager. In the present study, we demonstrated that LPS triggered global DNA and protein methylation in glomerular mesangial MES-13 cells. LPS upregulated protein expressions of enzymes central to cellular methylation reactions, especially protein arginine methyltransferase 6 (PRMT6) in MES-13 cells. Expression of the Klotho protein was diminished by LPS and was restored by 5-Aza-2'-deoxycytidine (5-Aza-2'-dc), AMI-1, and ammonium pyrrolidinedithiocarbamate (PDTC), but not adenosine aldehyde (AdOx). NF-κB was identified as a substrate for arginine methylation and interacted with PRMT6 in MES-13 cells. Inhibition of PRMT activity by AMI-1 blocked LPS-induced NF-κB nuclear translocation in MES-13 cells. Our data indicate that NF-κB negatively regulated Klotho expression with an interaction with PRMT6, which was upregulated by LPS in MES-13 cells.
Collapse
Affiliation(s)
- Kuen-Daw Tsai
- Department of Internal Medicine, China Medical University Beigang Hospital, Beigang Township, Yunlin County, Taiwan, Republic of China.,Institute of Molecular Biology, National Chung Cheng University, Minhsiung Township, Chiayi County, Taiwan, Republic of China.,School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan, Republic of China
| | - Wen-Xi Lee
- School of Biomedical Sciences, College of Medical Science and Technology, Chung Shan Medical University, Taichung, Taiwan, Republic of China.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan, Republic of China
| | - Wei Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chia-Yi Christian Hospital, Chiayi, Taiwan, Republic of China
| | - Bo-Yu Chen
- School of Biomedical Sciences, College of Medical Science and Technology, Chung Shan Medical University, Taichung, Taiwan, Republic of China.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan, Republic of China
| | - Kuan-Lin Chen
- School of Biomedical Sciences, College of Medical Science and Technology, Chung Shan Medical University, Taichung, Taiwan, Republic of China.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan, Republic of China
| | - Tzu-Chia Hsiao
- School of Biomedical Sciences, College of Medical Science and Technology, Chung Shan Medical University, Taichung, Taiwan, Republic of China.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan, Republic of China
| | - Sue-Hong Wang
- School of Biomedical Sciences, College of Medical Science and Technology, Chung Shan Medical University, Taichung, Taiwan, Republic of China.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan, Republic of China
| | - Yi-Ju Lee
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Shan-Yuan Liang
- School of Biomedical Sciences, College of Medical Science and Technology, Chung Shan Medical University, Taichung, Taiwan, Republic of China.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan, Republic of China
| | - Jia-Ching Shieh
- School of Biomedical Sciences, College of Medical Science and Technology, Chung Shan Medical University, Taichung, Taiwan, Republic of China.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan, Republic of China
| | - Ting-Hui Lin
- School of Biomedical Sciences, College of Medical Science and Technology, Chung Shan Medical University, Taichung, Taiwan, Republic of China.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan, Republic of China
| |
Collapse
|
61
|
Richter B, Faul C. FGF23 Actions on Target Tissues-With and Without Klotho. Front Endocrinol (Lausanne) 2018; 9:189. [PMID: 29770125 PMCID: PMC5940753 DOI: 10.3389/fendo.2018.00189] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/06/2018] [Indexed: 12/11/2022] Open
Abstract
Fibroblast growth factor (FGF) 23 is a phosphaturic hormone whose physiologic actions on target tissues are mediated by FGF receptors (FGFR) and klotho, which functions as a co-receptor that increases the binding affinity of FGF23 for FGFRs. By stimulating FGFR/klotho complexes in the kidney and parathyroid gland, FGF23 reduces renal phosphate uptake and secretion of parathyroid hormone, respectively, thereby acting as a key regulator of phosphate metabolism. Recently, it has been shown that FGF23 can also target cell types that lack klotho. This unconventional signaling event occurs in an FGFR-dependent manner, but involves other downstream signaling pathways than in "classic" klotho-expressing target organs. It appears that klotho-independent signaling mechanisms are only activated in the presence of high FGF23 concentrations and result in pathologic cellular changes. Therefore, it has been postulated that massive elevations in circulating levels of FGF23, as found in patients with chronic kidney disease, contribute to associated pathologies by targeting cells and tissues that lack klotho. This includes the induction of cardiac hypertrophy and fibrosis, the elevation of inflammatory cytokine expression in the liver, and the inhibition of neutrophil recruitment. Here, we describe the signaling and cellular events that are caused by FGF23 in tissues lacking klotho, and we discuss FGF23's potential role as a hormone with widespread pathologic actions. Since the soluble form of klotho can function as a circulating co-receptor for FGF23, we also discuss the potential inhibitory effects of soluble klotho on FGF23-mediated signaling which might-at least partially-underlie the pleiotropic tissue-protective functions of klotho.
Collapse
|
62
|
Słomiński B, Ryba-Stanisławowska M, Skrzypkowska M, Myśliwska J, Myśliwiec M. The KL-VS polymorphism of KLOTHO gene is protective against retinopathy incidence in patients with type 1 diabetes. Biochim Biophys Acta Mol Basis Dis 2017; 1864:758-763. [PMID: 29247834 DOI: 10.1016/j.bbadis.2017.12.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 12/04/2017] [Accepted: 12/11/2017] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND AIMS KLOTHO is an anti-ageing circulating hormone involved in insulin signaling, inflammation and vascular homeostasis through its protective effects on the endothelium and antioxidant actions. The common functional "KL-VS" variant of the KLOTHO gene is reproducibly associated with longevity in humans. Large number of studies have evaluated close relationship between KLOTHO protein and diabetes but the association between KL-VS variant and retinopathy in type 1 diabetes mellitus (T1D) is unknown. Therefore, in the present study we examined the association between the KL-VS polymorphism and the risk of diabetic retinopathy (DR) in patients with T1D. METHODS We examined 400 patients with T1D and 350 healthy age-matched controls. The analysis concerned KL-VS polymorphism along with the levels of serum inflammatory (CRP, TNF-α) and anti-inflammatory (IL-10) markers, pro-angiogenic (angiogenin) and anti-angiogenic interferon gamma-induced protein 10 (IP-10) factors as well as adhesion molecules (ICAM-1, ICAM-3). RESULTS We did not find significant association between T1D and KL-VS alleles. However, we observed that the incidence of KL-VS genotype is lower in a group with retinopathy in comparison to diabetic patients without this complication. Moreover, we established that KL-VS carriers had the lowest levels of inflammatory markers, pro-angiogenic factors and adhesion molecules. Simultaneously, the KL-VS carriers had increased serum levels of anti-inflammatory and anti-angiogenic cytokines than holders bearing wild type genotype. CONCLUSIONS In conclusion, the findings of our studies suggest that the functional KL-VS variant of the KLOTHO gene protects against the development of retinopathy in patients with T1D.
Collapse
Affiliation(s)
- Bartosz Słomiński
- Department of Immunology, Medical University of Gdańsk, Dębinki 1, 80-211 Gdańsk, Poland.
| | | | - Maria Skrzypkowska
- Department of Immunology, Medical University of Gdańsk, Dębinki 1, 80-211 Gdańsk, Poland
| | - Jolanta Myśliwska
- Department of Immunology, Medical University of Gdańsk, Dębinki 1, 80-211 Gdańsk, Poland
| | - Małgorzata Myśliwiec
- Chair & Clinics of Paediatrics, Diabetology and Endocrinology, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| |
Collapse
|
63
|
Jaturakan O, Buranakarl C, Dissayabutra T, Chaiyabutr N, Kijtawornrat A, Rungsipipat A. Changes of Klotho protein and Klotho mRNA expression in a hydroxy-L-proline induced hyperoxaluric rat model. J Vet Med Sci 2017; 79:1861-1869. [PMID: 28943597 PMCID: PMC5709566 DOI: 10.1292/jvms.17-0340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Klotho protein is recognized as having a renoprotective effect and is used as a biomarker for kidney injury. We investigated the level of Klotho protein in hyperoxaluria-induced kidney injury and the effects of vitamin E (Vit E)
and vitamin C (Vit C) supplementation. Hyperoxaluria was induced by feeding 2% (w/v) Hydroxy-L-proline (HLP) in the drinking water for 21 days. Rats were divided into 5 groups; control (Group 1, n=7), HLP treated rats that
received nothing else (Group 2, n=7), Vit E (Group 3, n=6), Vit C (Group 4, n=6) and both Vit E and Vit C (Group 5, n=7). Vit E (200 mg/kg) was injected on days 1, 6, 11 and 16, while Vit C (500 mg/kg) was given intravenously on
days 1 and 11. The Klotho protein levels and oxidative status were measured. The expression level of kidney Klotho protein expression was significantly reduced by HLP-treatment, while the mRNA expression was higher
(P<0.05), the plasma and kidney malondialdehyde and kidney superoxide dismutase activities were increased, and the kidney reduced glutathione and urinary total antioxidant status were decreased
(P<0.05). All of these changes were ameliorated by administration of Vit E, Vit C or especially the co-administration of both. In conclusion, HLP-induced hyperoxaluria reduced the kidney Klotho protein level,
which could be restored by Vit E and/or Vit C.
Collapse
Affiliation(s)
- Orapun Jaturakan
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Henri Dunant Rd., Pathumwan, Bangkok 10330, Thailand
| | - Chollada Buranakarl
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Henri Dunant Rd., Pathumwan, Bangkok 10330, Thailand
| | - Thasinas Dissayabutra
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| | - Narongsak Chaiyabutr
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Henri Dunant Rd., Pathumwan, Bangkok 10330, Thailand
| | - Anusak Kijtawornrat
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Henri Dunant Rd., Pathumwan, Bangkok 10330, Thailand
| | - Anudep Rungsipipat
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Henri Dunant Rd., Pathumwan, Bangkok 10330, Thailand
| |
Collapse
|
64
|
Abstract
The vertebrate endoskeleton is not a mere frame for muscle attachment to facilitate locomotion, but is a massive organ integrated with many physiologic functions including mineral and energy metabolism. Mineral balance is maintained by tightly controlled ion fluxes that are external (intestine and kidney) and internal (between bone and other organs), and are regulated and coordinated by many endocrine signals between these organs. The endocrine fibroblast growth factors (FGFs) and Klotho gene families are complex systems that co-evolved with the endoskeleton. In particular, FGF23 and αKlotho which are primarily derived from bone and kidney respectively, are critical in maintaining mineral metabolism where each of these proteins serving highly diverse roles; abound with many unanswered questions regarding their upstream regulation and downstream functions. Genetic lesions of components of this network produce discreet disturbances in many facets of mineral metabolism. One acquired condition with colossal elevations of FGF23 and suppression of αKlotho is chronic kidney disease where multiple organ dysfunction contributes to the morbidity and mortality. However, the single most important group of derangements that encompasses the largest breadth of complications is mineral metabolism disorders. Mineral metabolic disorders in CKD impact negatively and significantly on the progression of renal disease as well as extra-renal complications. Knowledge of the origin, nature, and impact of phosphate, FGF23, and αKlotho derangements is pivotal to understanding the pathophysiology and treatment of CKD.
Collapse
Affiliation(s)
- Makoto Kuro-O
- Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan; Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Orson W Moe
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
65
|
Neyra JA, Hu MC. Potential application of klotho in human chronic kidney disease. Bone 2017; 100:41-49. [PMID: 28115282 PMCID: PMC5474175 DOI: 10.1016/j.bone.2017.01.017] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 01/18/2017] [Accepted: 01/18/2017] [Indexed: 01/13/2023]
Abstract
The extracellular domain of transmembrane alpha-Klotho (αKlotho, hereinafter simply called Klotho) is cleaved by secretases and released into the circulation as soluble Klotho. Soluble Klotho in the circulation starts to decline early in chronic kidney disease (CKD) stage 2 and urinary Klotho possibly even earlier in CKD stage 1. Therefore soluble Klotho could serve as an early and sensitive marker of kidney function decline. Moreover, preclinical animal data support Klotho deficiency is not just merely a biomarker, but a pathogenic factor for CKD progression and extrarenal CKD complications including cardiovascular disease and disturbed mineral metabolism. Prevention of Klotho decline, re-activation of endogenous Klotho production or supplementation of exogenous Klotho are all associated with attenuation of renal fibrosis, retardation of CKD progression, improvement of mineral metabolism, amelioration of cardiomyopathy, and alleviation of vascular calcification in CKD. Therefore Klotho is not only a diagnostic and/or prognostic marker for CKD, but the treatment of Klotho deficiency may be a promising strategy to prevent, retard, and decrease the burden of comorbidity in CKD.
Collapse
Affiliation(s)
- Javier A Neyra
- Department of Internal Medicine, University of Texas Southwestern Medical Center, USA; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, USA
| | - Ming Chang Hu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, USA; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, USA.
| |
Collapse
|
66
|
Tan SJ, Smith ER, Holt SG, Hewitson TD, Toussaint ND. Soluble klotho may be a marker of phosphate reabsorption. Clin Kidney J 2017; 10:397-404. [PMID: 28616218 PMCID: PMC5466110 DOI: 10.1093/ckj/sfw146] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 12/05/2016] [Indexed: 12/20/2022] Open
Abstract
Background: Membrane-bound α-klotho functions as a co-receptor with fibroblast growth factor receptor at the renal tubule conferring specificity to fibroblast growth factor-23 (FGF-23), allowing it to inhibit tubular phosphate reabsorption at physiological concentrations. α-klotho also exists as a soluble protein. However, the complex interrelationships between soluble α-klotho (sKl), FGF-23 and phosphate reabsorption are poorly understood, with little known about the links between sKl, FGF-23 and phosphate reabsorption in chronic kidney disease (CKD). This study addresses this issue in a cohort of patients with and without CKD. Methods: We conducted a single-centre, cross-sectional study of contemporaneously obtained samples of blood and 24-h urine biochemistry along with sKl and intact FGF-23 (iFGF-23) from non-dialysis-dependent CKD patients and healthy volunteers. Pearson's correlation coefficients were used to determine correlations between natural log-transformed (Ln) sKl and iFGF-23 with other parameters of interest. Backward multivariate analysis was undertaken to evaluate the relationship between mineral parameters. Results: One hundred and sixteen participants (77 with CKD and 39 healthy volunteers) were studied, of which 74 (63.8%) were male. The median age was 61 (interquartile range 49-71) years. Those with CKD had lower sKl (408 versus 542 pg/mL), higher iFGF-23 (94 versus 41 pg/mL), higher fractional excretion of phosphate (25.05 versus 10.98%) and lower daily urinary phosphate excretion (UPE) (24.8 versus 32.3 mmol/L) compared with healthy volunteers (all P ≤ 0.002). Age correlated inversely and estimated glomerular filtration rate (eGFR) correlated positively with phosphate reabsorption and Ln(sKl), while the opposite was seen with Ln(iFGF23). Upon multivariate analysis, eGFR, Ln(sKl) and parathyroid hormone were independently associated with phosphate reabsorption, whereas Ln(iFGF-23) was not, after adjustment for age. Conclusions: Abnormalities in phosphate regulatory pathways are disturbed early in CKD. While iFGF-23 is associated with phosphate excretion on univariate analyses, sKl demonstrates a significant association with phosphate reabsorption independent of iFGF-23, and this relationship deserves further exploration.
Collapse
Affiliation(s)
- Sven-Jean Tan
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Australia
- Department of Medicine (RMH), University of Melbourne, Melbourne, Australia
| | - Edward R. Smith
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Australia
| | - Stephen G. Holt
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Australia
- Department of Medicine (RMH), University of Melbourne, Melbourne, Australia
| | - Tim D. Hewitson
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Australia
- Department of Medicine (RMH), University of Melbourne, Melbourne, Australia
| | - Nigel D. Toussaint
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Australia
- Department of Medicine (RMH), University of Melbourne, Melbourne, Australia
| |
Collapse
|
67
|
Dalton GD, Xie J, An SW, Huang CL. New Insights into the Mechanism of Action of Soluble Klotho. Front Endocrinol (Lausanne) 2017; 8:323. [PMID: 29250031 PMCID: PMC5715364 DOI: 10.3389/fendo.2017.00323] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/02/2017] [Indexed: 12/31/2022] Open
Abstract
The klotho gene encodes a type I single-pass transmembrane protein that contains a large extracellular domain, a membrane spanning segment, and a short intracellular domain. Klotho protein exists in several forms including the full-length membrane form (mKl) and a soluble circulating form [soluble klotho (sKl)]. mKl complexes with fibroblast growth factor receptors to form coreceptors for FGF23, which allows it to participate in FGF23-mediated signal transduction and regulation of phosphate and calcium homeostasis. sKl is present in the blood, urine, and cerebrospinal fluid where it performs a multitude of functions including regulation of ion channels/transporters and growth factor signaling. How sKl exerts these pleiotropic functions is poorly understood. One hurdle in understanding sKl's mechanism of action as a "hormone" has been the inability to identify a receptor that mediates its effects. In the body, the kidneys are a major source of sKl and sKl levels decline during renal disease. sKl deficiency in chronic kidney disease makes the heart susceptible to stress-induced injury. Here, we summarize the current knowledge of mKl's mechanism of action, the mechanistic basis of sKl's protective, FGF23-independent effects on the heart, and provide new insights into the mechanism of action of sKl focusing on recent findings that sKl binds sialogangliosides in membrane lipid rafts to regulate growth factor signaling.
Collapse
Affiliation(s)
- George D. Dalton
- Department of Medicine, Division of Gastroenterology, Duke University Medical Center, Durham, NC, United States
| | - Jian Xie
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Sung-Wan An
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Chou-Long Huang
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Iowa Carver College of Medicine, Iowa City, IA, United States
- *Correspondence: Chou-Long Huang,
| |
Collapse
|
68
|
Lu X, Hu MC. Klotho/FGF23 Axis in Chronic Kidney Disease and Cardiovascular Disease. KIDNEY DISEASES 2016; 3:15-23. [PMID: 28785560 DOI: 10.1159/000452880] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 10/26/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Membrane αKlotho (hereinafter called Klotho) is highly expressed in the kidney and functions as a coreceptor of FGF receptors (FGFRs) to activate specific fibroblast growth factor 23 (FGF23) signal pathway. FGF23 is produced in bones and participates in the maintenance of mineral homeostasis. The extracellular domain of transmembrane Klotho can be cleaved by secretases and released into the circulation as soluble Klotho. Soluble Klotho does not only weakly activate FGFRs to transduce the FGF23 signaling pathway, but also functions as an enzyme and hormonal substance to play a variety of biological functions. FGF23 exerts its biological effects through activation of FGFRs in a Klotho-dependent manner. However, extremely high FGF23 can exert its pathological action in a Klotho-independent manner. SUMMARY The decline in serum and urinary Klotho followed by a rise in serum FGF23 at an early stage of chronic kidney disease (CKD) functions as an early biomarker for kidney dysfunction and can also serve as a predictor for risk of cardiovascular disease (CVD) and mortality in both CKD patients and the general population. Moreover, Klotho deficiency is a pathogenic factor for CKD progression and CVD. FGF23 may also contribute to CVD. Prevention of Klotho decline, reactivation of endogenous Klotho production, or supplementation of exogenous Klotho attenuate renal fibrosis, retard CKD progression, improve mineral metabolism, ameliorate cardiomyopathy, and alleviate vascular calcification in CKD. However, the poor CVD outcome after depletion of FGF23 with FGF23 antibody stimulates the generation of a more specific inhibitor of FGF23 for CKD treatment. KEY MESSAGE Klotho/FGF23 may not only be diagnostic and/or prognostic biomarkers for CKD and CVD, but are also pathogenic contributors to CKD progression and CVD development. The Klotho/FGF23 axis should be a novel target for renal clinics.
Collapse
Affiliation(s)
- Xiang Lu
- Department of Geriatrics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, PR China
| | - Ming Chang Hu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
69
|
Kim SS, Song SH, Kim IJ, Lee EY, Lee SM, Chung CH, Kwak IS, Lee EK, Kim YK. Decreased plasma α-Klotho predict progression of nephropathy with type 2 diabetic patients. J Diabetes Complications 2016; 30:887-92. [PMID: 27037042 DOI: 10.1016/j.jdiacomp.2016.03.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 03/04/2016] [Accepted: 03/09/2016] [Indexed: 01/07/2023]
Abstract
AIM The potential role of soluble α-klotho in diabetic kidney disease has not yet been evaluated. The aim of this study was to evaluate the association of plasma and/or urine α-klotho with the progression of type 2 diabetic nephropathy. METHODS The baseline values of plasma and urine α-klotho were measured in 147 patients with type 2 diabetes mellitus with an estimated glomerular filtration rate (eGFR) of ≥60mL/min/1.73m(2). In this prospective observational study, a total of 109 type 2 diabetic patients were followed up for 34months (8-50 months). RESULTS Plasma α-klotho, but not urine α-klotho, was negatively correlated with the decline of eGFR (r=-0.304, P=0.001; r=0.042, P=0.068, respectively). After adjusting for several clinical parameters, baseline eGFR and urine ACR, plasma α-klotho was significantly associated with the decline of eGFR (r=-0.219, P=0.008). In the normoalbuminuria group (n=63), the plasma α-klotho remained significantly associated with a decline in eGFR (r=0.324, P=0.004) in the final model. CONCLUSIONS It is suggested that plasma α-klotho may be an early biomarker for predicting renal impairment in type 2 diabetic patients. The disappearance of a compensatory increase of plasma α-klotho might be a predictive marker for the progression of type 2 diabetic nephropathy.
Collapse
Affiliation(s)
- Sang Soo Kim
- Department of Internal Medicine, Pusan National University Hospital, Busan, Korea; Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Sang Heon Song
- Department of Internal Medicine, Pusan National University Hospital, Busan, Korea; Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - In Joo Kim
- Department of Internal Medicine, Pusan National University Hospital, Busan, Korea; Biomedical Research Institute, Pusan National University Hospital, Busan, Korea.
| | - Eun Young Lee
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea.
| | - Su Mi Lee
- Department of Internal Medicine, Dong-A University Hospital, Busan, Korea
| | - Choon Hee Chung
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Ihm Soo Kwak
- Department of Internal Medicine, Pusan National University Hospital, Busan, Korea
| | | | - Yong Ki Kim
- Kim Yong Ki Internal Medicine Clinic, Busan, Korea
| |
Collapse
|
70
|
Tan SJ, Smith ER, Hewitson TD, Holt SG, Toussaint ND. The importance of klotho in phosphate metabolism and kidney disease. Nephrology (Carlton) 2016; 19:439-49. [PMID: 24750549 DOI: 10.1111/nep.12268] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2014] [Indexed: 12/19/2022]
Abstract
The discovery of fibroblast growth factor-23 (FGF23) and its co-receptor α-klotho has broadened our understanding of mineral metabolism and led to a renewed research focus on phosphate homeostatic pathways in kidney disease. Expanding knowledge of these mechanisms, both in normal physiology and in pathology, identifies targets for potential interventions designed to reduce the complications of renal disease, particularly the cardiovascular sequelae. FGF23 has emerged as a major α-klotho-dependent endocrine regulator of mineral metabolism, functioning to activate vitamin D and as a phosphatonin. However, increasingly there is an appreciation that klotho may act independently as a phosphate regulator, as well as having significant activity in other key biological processes. This review outlines our current understanding of klotho, and its potential contribution to kidney disease and cardiovascular health.
Collapse
Affiliation(s)
- Sven-Jean Tan
- Department of Nephrology, The Royal Melbourne Hospital, Melbourne, Victoria, Australia; Department of Medicine (RMH), The University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | | | |
Collapse
|
71
|
Impact of Renal Transplantation and Nephrectomy on Urinary Soluble Klotho Protein. Transplant Proc 2016; 47:1697-9. [PMID: 26293036 DOI: 10.1016/j.transproceed.2015.06.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 06/06/2015] [Accepted: 06/16/2015] [Indexed: 12/29/2022]
Abstract
BACKGROUND Klotho is a single-pass transmembrane protein predominantly expressed in the kidneys. The soluble form of klotho has been shown to participate in various pathophysiological activities. However, information regarding the kinetics of soluble klotho remains limited. We herein assessed serial changes in the amounts of 24-hour urinary excreted soluble klotho among renal transplant recipients and concomitant living donors before and after transplantation. METHODS A total of 15 recipients and donors were included in the current study, and the amounts of urinary soluble klotho were quantified using a sandwich enzyme-linked immunosorbent assay. RESULTS Urine samples were available in 6 of the 15 recipients prior to the procedure. The amounts of urinary klotho in these 6 recipients and overall living donors at the baseline were 58.6 ng/day (IR: 29.3-142) and 698.8 ng/day (IR: 62.3-1619.5), respectively. Those in the recipients on postoperative day 2 (median 522.3 ng/day; IR 337.1-1168.5, P < .05) and day 5 (median 723.2 ng/day; IR 254.7-1238.6, P < .05) were significantly higher than the baseline values. Among the living donors, only a transient increase was observed in the amounts of urinary klotho on postoperative day 2. CONCLUSION The current data regarding the urinary soluble klotho in recipients support the hypothesis that the kidney is a major source of urinary soluble klotho among the numerous components of the urinary tract. In living donors, the complex nature of events associated with acute reductions in the renal mass may modulate the release of soluble klotho from the kidneys into the urine.
Collapse
|
72
|
Abstract
Alpha-Klotho (αKlotho) protein is encoded by the gene, Klotho, and functions as a coreceptor for endocrine fibroblast growth factor-23. The extracellular domain of αKlotho is cleaved by secretases and released into the circulation where it is called soluble αKlotho. Soluble αKlotho in the circulation starts to decline in chronic kidney disease (CKD) stage 2 and urinary αKlotho in even earlier CKD stage 1. Therefore soluble αKlotho is an early and sensitive marker of decline in kidney function. Preclinical data from numerous animal experiments support αKlotho deficiency as a pathogenic factor for CKD progression and extrarenal CKD complications including cardiac and vascular disease, hyperparathyroidism, and disturbed mineral metabolism. αKlotho deficiency induces cell senescence and renders cells susceptible to apoptosis induced by a variety of cellular insults including oxidative stress. αKlotho deficiency also leads to defective autophagy and angiogenesis and promotes fibrosis in the kidney and heart. Most importantly, prevention of αKlotho decline, upregulation of endogenous αKlotho production, or direct supplementation of soluble αKlotho are all associated with attenuation of renal fibrosis, retardation of CKD progression, improvement of mineral metabolism, amelioration of cardiac function and morphometry, and alleviation of vascular calcification in CKD. Therefore in rodents, αKlotho is not only a diagnostic and prognostic marker for CKD but the enhancement of endogenous or supplement of exogenous αKlotho are promising therapeutic strategies to prevent, retard, and decrease the comorbidity burden of CKD.
Collapse
Affiliation(s)
- J A Neyra
- University of Texas Southwestern Medical Center, Dallas, TX, United States; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - M C Hu
- University of Texas Southwestern Medical Center, Dallas, TX, United States; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
73
|
Hu MC, Shi M, Zhang J, Addo T, Cho HJ, Barker SL, Ravikumar P, Gillings N, Bian A, Sidhu SS, Kuro-o M, Moe OW. Renal Production, Uptake, and Handling of Circulating αKlotho. J Am Soc Nephrol 2016; 27:79-90. [PMID: 25977312 PMCID: PMC4696570 DOI: 10.1681/asn.2014101030] [Citation(s) in RCA: 205] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 03/14/2015] [Indexed: 01/13/2023] Open
Abstract
αKlotho is a multifunctional protein highly expressed in the kidney. Soluble αKlotho is released through cleavage of the extracellular domain from membrane αKlotho by secretases to function as an endocrine/paracrine substance. The role of the kidney in circulating αKlotho production and handling is incompletely understood, however. Here, we found higher αKlotho concentration in suprarenal compared with infrarenal inferior vena cava in both rats and humans. In rats, serum αKlotho concentration dropped precipitously after bilateral nephrectomy or upon treatment with inhibitors of αKlotho extracellular domain shedding. Furthermore, the serum half-life of exogenous αKlotho in anephric rats was four- to five-fold longer than that in normal rats, and exogenously injected labeled recombinant αKlotho was detected in the kidney and in urine of rats. Both in vivo (micropuncture) and in vitro (proximal tubule cell line) studies showed that αKlotho traffics from the basal to the apical side of the proximal tubule via transcytosis. Thus, we conclude that the kidney has dual roles in αKlotho homeostasis, producing and releasing αKlotho into the circulation and clearing αKlotho from the blood into the urinary lumen.
Collapse
Affiliation(s)
- Ming Chang Hu
- Departments of Internal Medicine, Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas;
| | - Mingjun Shi
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | | | | | - Han Ju Cho
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sarah L Barker
- Banting and Best Department of Medical Research and Department of Molecular Genetics, The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada; and
| | - Priya Ravikumar
- Departments of Internal Medicine, Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Nancy Gillings
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ao Bian
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sachdev S Sidhu
- Banting and Best Department of Medical Research and Department of Molecular Genetics, The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada; and
| | - Makoto Kuro-o
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas; Pathology, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Orson W Moe
- Departments of Internal Medicine, Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas; Physiology, and
| |
Collapse
|
74
|
Massó A, Sánchez A, Gimenez-Llort L, Lizcano JM, Cañete M, García B, Torres-Lista V, Puig M, Bosch A, Chillon M. Secreted and Transmembrane αKlotho Isoforms Have Different Spatio-Temporal Profiles in the Brain during Aging and Alzheimer's Disease Progression. PLoS One 2015; 10:e0143623. [PMID: 26599613 PMCID: PMC4658185 DOI: 10.1371/journal.pone.0143623] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 11/06/2015] [Indexed: 12/28/2022] Open
Abstract
The Klotho protein is a β-glucuronidase, and its overexpression is associated with life extension. Its mechanism of action is not fully understood, although it has been recently reported that αKlotho improves synaptic and cognitive functions, and it may also influence a variety of structures and functions during CNS maturation and aging. The αKlotho gene has two transcripts, one encoding a transmembrane isoform (m-KL), and the other a putative secreted isoform (s-KL). Unfortunately, little is known about the secreted αKlotho isoform, since available antibodies cannot discriminate s-KL from the KL1 domain cleaved from the transmembrane isoform. This study shows, for the first time, that the klotho transcript produced by alternative splicing generates a stable protein (70 kDa), and that in contrast to the transmembrane Klotho isoform, it is ten times more abundant in the brain than in the kidney suggesting that the two isoforms may have different functions. We also studied whether klotho expression in the CNS was influenced by aging, Alzheimer's disease (AD), or a healthy lifestyle, such as voluntary moderate continuous exercise. We observed a strong correlation between high expression levels of the two klotho transcripts and the healthy status of the animals. Expression of Klotho in brain areas decayed more rapidly in the 3xTg-AD model of AD than in healthy animals, whilst moderate continuous exercise in adulthood prevents the decline in expression of both klotho transcripts.
Collapse
Affiliation(s)
- Anna Massó
- Departament Bioquímica i Biologia Molecular, Universitat Autònoma Barcelona, Bellaterra, Spain
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma Barcelona, Bellaterra, Spain
| | - Angela Sánchez
- Departament Bioquímica i Biologia Molecular, Universitat Autònoma Barcelona, Bellaterra, Spain
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma Barcelona, Bellaterra, Spain
| | - Lydia Gimenez-Llort
- Institut de Neurociencies, Universitat Autònoma Barcelona, Bellaterra, Spain
- Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Jose Miguel Lizcano
- Departament Bioquímica i Biologia Molecular, Universitat Autònoma Barcelona, Bellaterra, Spain
- Institut de Neurociencies, Universitat Autònoma Barcelona, Bellaterra, Spain
| | - Manuel Cañete
- Departament Bioquímica i Biologia Molecular, Universitat Autònoma Barcelona, Bellaterra, Spain
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma Barcelona, Bellaterra, Spain
| | - Belen García
- Departament Bioquímica i Biologia Molecular, Universitat Autònoma Barcelona, Bellaterra, Spain
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma Barcelona, Bellaterra, Spain
| | - Virginia Torres-Lista
- Institut de Neurociencies, Universitat Autònoma Barcelona, Bellaterra, Spain
- Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Meritxell Puig
- Departament Bioquímica i Biologia Molecular, Universitat Autònoma Barcelona, Bellaterra, Spain
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma Barcelona, Bellaterra, Spain
| | - Assumpció Bosch
- Departament Bioquímica i Biologia Molecular, Universitat Autònoma Barcelona, Bellaterra, Spain
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma Barcelona, Bellaterra, Spain
| | - Miguel Chillon
- Departament Bioquímica i Biologia Molecular, Universitat Autònoma Barcelona, Bellaterra, Spain
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma Barcelona, Bellaterra, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- * E-mail:
| |
Collapse
|
75
|
Adema AY, Vervloet MG, Blankenstein MA, Heijboer AC. α-Klotho is unstable in human urine. Kidney Int 2015; 88:1442-1444. [PMID: 26244922 DOI: 10.1038/ki.2015.238] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 05/28/2015] [Accepted: 06/04/2015] [Indexed: 11/09/2022]
Abstract
α-Klotho is an interesting new biomarker in kidney and cardiovascular disease. As α-Klotho is primarily expressed in renal epithelial tissue, various papers have reported α-Klotho concentrations in urine. As these studies did not address the reliability of urinary α-Klotho measurements and as urine is known to be a complex milieu, we investigated the stability of α-Klotho in both fresh catheter and fresh voided urine. α-Klotho was measured in these fresh urine samples and in the same samples under several other conditions. Storage of fresh catheter urine for 3 h at 37 °C, comparable to storage in the bladder, led to a 82% decrease in α-Klotho concentrations. Compared with fresh voided urine, α-Klotho concentrations decreased on an average of 45 and 82% after storage at -80 °C and -20 °C, respectively. An additional freeze-thaw cycle further decreased α-Klotho concentrations. The addition of a protease inhibitor or 0.1% albumin partly prevented degradation in fresh voided urine. Thus, α-Klotho is highly unstable in urine. Future studies showing results of urinary α-Klotho should mention conservation conditions and prove the reliability of the α-Klotho measurements.
Collapse
Affiliation(s)
- Aaltje Y Adema
- Department of Nephrology, VU University Medical Center, Amsterdam, The Netherlands
| | - Marc G Vervloet
- Department of Nephrology, VU University Medical Center, Amsterdam, The Netherlands.,Institute for Cardiovascular Research VU (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands
| | - Marinus A Blankenstein
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands
| | - Annemieke C Heijboer
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
76
|
Olauson H, Vervloet MG, Cozzolino M, Massy ZA, Ureña Torres P, Larsson TE. New insights into the FGF23-Klotho axis. Semin Nephrol 2015; 34:586-97. [PMID: 25498378 DOI: 10.1016/j.semnephrol.2014.09.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Abnormal mineral metabolism is a hallmark in patients with advanced chronic kidney disease (CKD). Hyperphosphatemia, and the homeostatic mechanisms controlling phosphate metabolism, have received particular attention over the past decade. The phosphate-regulating hormone fibroblast growth factor-23 (FGF23) was discovered through studies of rare hypophosphatemic disorders, whereas Klotho, which subsequently turned out to be a co-receptor for FGF23, was identified in a mouse model showing hyperphosphatemia and multiple aging-like traits. The FGF23-Klotho endocrine axis is a pivotal regulator of mineral metabolism. In CKD, early onset of Klotho deficiency contributes to renal FGF23 resistance and a maladaptive increase in circulating FGF23. FGF23 is an early biomarker of renal injury and increased FGF23 predicts adverse clinical outcomes, in particular cardiovascular disease. A paradigm of FGF23 excess and Klotho deficiency is proposed, in which FGF23 preferentially stimulates left ventricular hypertrophy, and loss of Klotho augments fibrosis, endothelial dysfunction, and vascular calcification. The clinical benefit of FGF23 and Klotho measurements remain uncertain, nevertheless, the FGF23-Klotho axis is a solid candidate for a novel diagnostic and therapeutic target in CKD.
Collapse
Affiliation(s)
- Hannes Olauson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Marc G Vervloet
- Department of Nephrology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Mario Cozzolino
- Department of Health Sciences, University of Milan, Renal Division, San Paolo Hospital, Milan, Italy
| | - Ziad A Massy
- Inserm U-1088, Université de Picardie Jules Verne (UPJV), Amiens, France; Division of Nephrology, Ambroise Paré Hospital, Paris-Ile-de-France-Ouest University (Université de Versailles Saint-Quentin-en-Yvelines (UVSQ)), Paris-Boulogne Billancourt, France
| | - Pablo Ureña Torres
- Service of Nephrology and Dialysis, Clinique du Landy and Department of Renal Physiology, Necker Hospital, University of Paris Descartes, Paris, France
| | - Tobias E Larsson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden; Department of Nephrology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
77
|
Otani-Takei N, Masuda T, Akimoto T, Honma S, Watanabe Y, Shiizaki K, Miki T, Kusano E, Asano Y, Kuro-o M, Nagata D. Association between Serum Soluble Klotho Levels and Mortality in Chronic Hemodialysis Patients. Int J Endocrinol 2015; 2015:406269. [PMID: 26604925 PMCID: PMC4641198 DOI: 10.1155/2015/406269] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 10/04/2015] [Accepted: 10/13/2015] [Indexed: 01/25/2023] Open
Abstract
Klotho is a single-pass transmembrane protein predominantly expressed in the kidney. The extracellular domain of Klotho is subject to ectodomain shedding and is released into the circulation as a soluble form. Soluble Klotho is also generated from alternative splicing of the Klotho gene. In mice, defects in Klotho expression lead to complex phenotypes resembling those observed in dialysis patients. However, the relationship between the level of serum soluble Klotho and overall survival in hemodialysis patients, who exhibit a state of Klotho deficiency, remains to be delineated. Here we prospectively followed a cohort of 63 patients with a mean duration of chronic hemodialysis of 6.7 ± 5.4 years for a median of 65 months. Serum soluble Klotho was detectable in all patients (median 371 pg/mL, interquartile range 309-449). Patients with serum soluble Klotho levels below the lower quartile (<309 pg/mL) had significantly higher cardiovascular and all-cause mortality rates. Furthermore, the higher all-cause mortality persisted even after adjustment for confounders (hazard ratio 4.14, confidence interval 1.29-13.48). We conclude that there may be a threshold for the serum soluble Klotho level associated with a higher risk of mortality.
Collapse
Affiliation(s)
- Naoko Otani-Takei
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Takahiro Masuda
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
- *Takahiro Masuda:
| | - Tetsu Akimoto
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Sumiko Honma
- Department of Nephrology, Japanese Red Cross Koga Hospital, Koga, Ibaraki, Japan
| | - Yuko Watanabe
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Kazuhiro Shiizaki
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Takuya Miki
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Eiji Kusano
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Yasushi Asano
- Department of Nephrology, Japanese Red Cross Koga Hospital, Koga, Ibaraki, Japan
| | - Makoto Kuro-o
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Daisuke Nagata
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| |
Collapse
|
78
|
Rotondi S, Pasquali M, Tartaglione L, Muci ML, Mandanici G, Leonangeli C, Sales S, Farcomeni A, Mazzaferro S. Soluble α -Klotho Serum Levels in Chronic Kidney Disease. Int J Endocrinol 2015; 2015:872193. [PMID: 25873958 PMCID: PMC4383388 DOI: 10.1155/2015/872193] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 11/17/2014] [Indexed: 02/08/2023] Open
Abstract
Transmembrane α-Klotho (TM-Klotho), expressed in renal tubules, is a cofactor for FGF23-receptor. Circulating soluble-α-Klotho (s-Klotho) results from TM-Klotho shedding and acts on Phosphate (P) and Calcium (Ca) tubular transport. Decreased TM-Klotho, described in experimental chronic kidney disease (CKD), prevents actions of FGF23 and lessens circulating s-Klotho. Thus, levels of s-Klotho could represent a marker of CKD-MBD. To evaluate the clinical significance of s-Klotho in CKD we assayed serum s-Klotho and serum FGF23 in 68 patients (age 58 ± 15; eGFR 45 ± 21 mL/min). s-Klotho was lower than normal (519 ± 183 versus 845 ± 330 pg/mL, P < .0001) in renal patients and its reduction was detectable since CKD stage 2 (P < .01). s-Klotho correlated positively with eGFR and serum calcium (Cas) and negatively with serum phosphate (Ps), PTH and FGF23. FGF23 was higher than normal (73 ± 51 versus 36 ± 11, P < .0002) with significantly increased levels since CKD stage 2 (P < .001). Our data indicate a negative effect of renal disease on circulating s-Klotho starting very early in CKD. Assuming that s-Klotho mirrors TM-Klotho synthesis, low circulating s-Klotho seems to reflect the ensuing of tubular resistance to FGF23, which, accordingly, is increased. We endorse s-Klotho as an early marker of CKD-MBD.
Collapse
Affiliation(s)
- Silverio Rotondi
- Department of Cardiovascular, Respiratory, Nephrology, Geriatric, and Anesthetic Sciences, “Sapienza” University, 5 Piazzale Aldo Moro, 00185 Rome, Italy
| | - Marzia Pasquali
- Department of Cardiovascular, Respiratory, Nephrology, Geriatric, and Anesthetic Sciences, “Sapienza” University, 5 Piazzale Aldo Moro, 00185 Rome, Italy
| | - Lida Tartaglione
- Department of Cardiovascular, Respiratory, Nephrology, Geriatric, and Anesthetic Sciences, “Sapienza” University, 5 Piazzale Aldo Moro, 00185 Rome, Italy
| | - Maria Luisa Muci
- Department of Cardiovascular, Respiratory, Nephrology, Geriatric, and Anesthetic Sciences, “Sapienza” University, 5 Piazzale Aldo Moro, 00185 Rome, Italy
| | - Giusy Mandanici
- Department of Cardiovascular, Respiratory, Nephrology, Geriatric, and Anesthetic Sciences, “Sapienza” University, 5 Piazzale Aldo Moro, 00185 Rome, Italy
| | - Cristiana Leonangeli
- Department of Cardiovascular, Respiratory, Nephrology, Geriatric, and Anesthetic Sciences, “Sapienza” University, 5 Piazzale Aldo Moro, 00185 Rome, Italy
| | - Silvia Sales
- Department of Cardiovascular, Respiratory, Nephrology, Geriatric, and Anesthetic Sciences, “Sapienza” University, 5 Piazzale Aldo Moro, 00185 Rome, Italy
| | - Alessio Farcomeni
- Department of Public Health and Infectious Diseases, Section of Statistics, “Sapienza” University, 5 Piazzale Aldo Moro, 00185 Rome, Italy
| | - Sandro Mazzaferro
- Department of Cardiovascular, Respiratory, Nephrology, Geriatric, and Anesthetic Sciences, “Sapienza” University, 5 Piazzale Aldo Moro, 00185 Rome, Italy
- *Sandro Mazzaferro:
| |
Collapse
|
79
|
Buiten MS, de Bie MK, Bouma-de Krijger A, van Dam B, Dekker FW, Jukema JW, Rabelink TJ, Rotmans JI. Soluble Klotho is not independently associated with cardiovascular disease in a population of dialysis patients. BMC Nephrol 2014; 15:197. [PMID: 25495997 PMCID: PMC4293085 DOI: 10.1186/1471-2369-15-197] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 11/24/2014] [Indexed: 01/08/2023] Open
Abstract
Background Dialysis patients suffer from a high burden of cardiovascular disease (CVD). Partly this is due to progressive deterioration of calcium-phosphate homeostasis. Previous studies suggested that besides FGF-23, low levels of Klotho, a protein linked to aging, might constitute a key factor in this detrimental relationship. The purpose of the present study was to determine the relationship between serum Klotho (sKlotho) and the presence of CVD in dialysis patients. Methods Plasma levels of sKlotho were measured in a cohort of dialysis patients and related to left ventricular (LV) dysfunction (defined as a LV ejection fraction <45%) and LV mass using echocardiography. Coronary artery disease (CAD) and calcification score were assessed using computed tomography angiography. Abdominal aortic calcification score (AACscore) was measured by abdominal X-ray. Results We included 127 dialysis patients, 67 ± 7 years old, 76% male, 67% on hemodialysis, median sKlotho 460 pg/mL (25th-75th percentile 350-620 pg/mL). Patients with a low sKlotho (<460 pg/mL) showed significantly more CAD (81% versus 61%; p = 0.02) and LV dysfunction (19% versus 3%; p < 0.01). However, after adjusting for confounders, sKlotho was not independently associated with the presence of CVD or the AACscore. Conclusions In the present cohort of dialysis patients, sKlotho was not independently associated with CVD. However, patients with a low sKlotho level (<460 pg/mL) did show CAD and LV dysfunction more frequently. Therefore, while sKlotho might be a marker for CVD in dialysis patients, the current data does not support a direct cardioprotective effect of sKlotho.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Joris I Rotmans
- Department of Nephrology, Leiden University Medical Center, P O Box 9600, 2300 RC Leiden, The Netherlands.
| |
Collapse
|
80
|
Feger M, Mia S, Pakladok T, Nicolay JP, Alesutan I, Schneider SW, Voelkl J, Lang F. Down-regulation of renal klotho expression by Shiga toxin 2. Kidney Blood Press Res 2014; 39:441-9. [PMID: 25471359 DOI: 10.1159/000368457] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Shiga toxin 2 may trigger classical hemolytic uremic syndrome (HUS) eventually leading to renal failure. Klotho, a transmembrane protein, protease and hormone mainly expressed in kidney is involved in the regulation of renal phosphate excretion and also retains renal protective effects. Renal failure is associated with renal depletion of klotho. The present study explored the influence of Shiga toxin 2 on renal klotho expression. METHODS Mice were injected with either solvent or Shiga toxin 2 and urinary flow rate and phosphate excretion were determined in metabolic cages. Renal transcript levels were measured by quantitative RT-PCR and renal protein abundance by Western blotting. Plasma concentrations of 1,25(OH)2D3 and FGF23 were determined by ELISA and plasma phosphate and urea concentrations by photometry. RESULTS Shiga toxin 2 treatment was followed by increase of plasma urea concentration, urinary flow rate and renal phosphate excretion but not of plasma phosphate concentration. Shiga toxin 2 treatment strongly decreased klotho mRNA expression and klotho protein abundance in renal tissue. Shiga toxin 2 treatment further increased tumor necrosis factor (Tnfα) mRNA levels, as well as protein abundance of phosphorylated p38 MAPK in renal tissue. The treatment significantly increased renal Cyp27b1 and decreased renal Cyp24a1 mRNA levels without significantly altering plasma 1,25(OH)2D3 levels. Shiga toxin 2 treatment was further followed by increase of plasma FGF23 concentrations. CONCLUSION Shiga toxin 2 treatment stimulated Tnfα transcription, down-regulated renal klotho expression and increased FGF23 formation, effects presumably contributing to renal tissue injury.
Collapse
Affiliation(s)
- Martina Feger
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
81
|
Nitta K, Nagano N, Tsuchiya K. Fibroblast growth factor 23/klotho axis in chronic kidney disease. Nephron Clin Pract 2014; 128:1-10. [PMID: 25402964 DOI: 10.1159/000365787] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 06/26/2014] [Indexed: 12/27/2022] Open
Abstract
Fibroblast growth factor-23 (FGF23) is a bone-derived hormone that regulates phosphate and 1,25-hydroxyvitamin D [1,25(OH)2D] metabolism. FGF23 binds to FGF receptor 1 with its coreceptor Klotho and maintains serum phosphate levels within the normal range by increasing renal phosphate excretion. In addition, FGF23 reduces the synthesis and accelerates the degradation of 1,25(OH)2D to reduce intestinal phosphate absorption. Moreover, FGF23 acts at the parathyroid gland to decrease parathyroid hormone synthesis and secretion. In chronic kidney disease (CKD), serum FGF23 levels rise exponentially as renal function declines long before a significant increase in serum phosphate concentration occurs. Although there is room for argument, FGF23 and Klotho are recently reported contributors to vascular calcification. Finally, prospective observational studies have shown that serum FGF23 concentrations predict mortality not only among dialysis patients but among predialysis CKD patients. In addition to being a coreceptor for FGF23, Klotho circulates as an endocrine substance and exerts a multitude of effects. This review describes recent advances in research on the FGF23-Klotho axis in CKD. © 2014 S. Karger AG, Basel.
Collapse
Affiliation(s)
- Kosaku Nitta
- Kidney Center, Department of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | | | | |
Collapse
|
82
|
Barker SL, Pastor J, Carranza D, Quiñones H, Griffith C, Goetz R, Mohammadi M, Ye J, Zhang J, Hu MC, Kuro-o M, Moe OW, Sidhu SS. The demonstration of αKlotho deficiency in human chronic kidney disease with a novel synthetic antibody. Nephrol Dial Transplant 2014; 30:223-33. [PMID: 25324355 DOI: 10.1093/ndt/gfu291] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND αKlotho is the prototypic member of the Klotho family and is most highly expressed in the kidney. αKlotho has pleiotropic biologic effects, and in the kidney, its actions include regulation of ion transport, cytoprotection, anti-oxidation and anti-fibrosis. In rodent models of chronic kidney disease (CKD), αKlotho deficiency has been shown to be an early biomarker as well as a pathogenic factor. The database for αKlotho in human CKD remains controversial even after years of study. METHODS We used a synthetic antibody library to identify a high-affinity human antigen-binding fragment that recognizes human, rat and mouse αKlotho primarily in its native, rather than denatured, form. RESULTS Using an immunoprecipitation-immunoblot (IP-IB) assay, we measured both serum and urinary levels of full-length soluble αKlotho in humans and established that human CKD is associated with αKlotho deficiency in serum and urine. αKlotho levels were detectably lower in early CKD preceding disturbances in other parameters of mineral metabolism and progressively declined with CKD stages. We also found that exogenously added αKlotho is inherently unstable in the CKD milieu suggesting that decreased production may not be the sole reason for αKlotho deficiency. CONCLUSION Synthetic antibody libraries harbor tremendous potential for a variety of biomedical and clinical applications. Using such a reagent, we furnish data in support of αKlotho deficiency in human CKD, and we set the foundation for the development of diagnostic and therapeutic applications of anti-αKlotho antibodies.
Collapse
Affiliation(s)
- Sarah L Barker
- Banting and Best Department of Medical Research and Department of Molecular Genetics, The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Johanne Pastor
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Danielle Carranza
- Banting and Best Department of Medical Research and Department of Molecular Genetics, The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Henry Quiñones
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Carolyn Griffith
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Regina Goetz
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Moosa Mohammadi
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Jianfeng Ye
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jianning Zhang
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ming Chang Hu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Makoto Kuro-o
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Orson W Moe
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sachdev S Sidhu
- Banting and Best Department of Medical Research and Department of Molecular Genetics, The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
83
|
Hage V, Pelletier S, Dubourg L, Drai J, Cuerq C, Lemoine S, Hadj-Aissa A, Laville M, Fouque D. In chronic kidney disease, serum α-Klotho is related to serum bicarbonate and proteinuria. J Ren Nutr 2014; 24:390-4. [PMID: 25193108 DOI: 10.1053/j.jrn.2014.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/14/2014] [Accepted: 06/24/2014] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Klotho is an "aging-suppressor" gene and encodes a single-pass transmembrane protein predominantly expressed in renal tubules. Whether chronic kidney disease (CKD) affects serum Klotho is poorly documented. We aimed to measure the relationship of serum α-Klotho with renal function, acid-base status, bone biomarkers, and proteinuria in CKD patients. DESIGN SETTING, PARTICIPANTS, AND MEASUREMENTS We measured serum α-Klotho, serum FGF23, and glomerular filtration rate by inulin clearance in 60 CKD patients between January and July 2011. We also measured serum creatinine, bicarbonate, calcium, phosphorus, parathyroid hormone, C-reactive protein, and 25-OH vitamin D. Proteinuria was obtained from a 24-h urine collection. RESULTS The median serum α-Klotho was 478 (348-658) pg/mL. We found an inverse relationship between serum α-Klotho and serum creatinine (r = -0.36, P = .007), proteinuria (r = -0.36, P = .013), and a positive relationship with serum bicarbonate (r = 0.33, P = .011). There was no further significant relation between serum α-Klotho and inulin clearance or serum FGF23. Multiple regression analysis including serum bicarbonate, serum creatinine, and proteinuria indicated that only serum bicarbonate was associated with serum α-Klotho (P = .003). CONCLUSIONS This study shows that in CKD, serum α-Klotho is related to serum bicarbonate and proteinuria and not to renal function. Further research is required to determine whether correcting these 2 amenable conditions would improve serum α-Klotho.
Collapse
Affiliation(s)
- Valerie Hage
- Département de Néphrologie-Dialyse-Nutrition, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Solenne Pelletier
- Département de Néphrologie-Dialyse-Nutrition, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre Bénite, France; Service d'Explorations Fonctionnelles Rénales et Métaboliques, Hôpital Edouard Herriot, Lyon, France; INSERM U1033, Hôpital Edouard HERRIOT, Lyon, France
| | - Laurence Dubourg
- Service d'Explorations Fonctionnelles Rénales et Métaboliques, Hôpital Edouard Herriot, Lyon, France
| | - Jocelyne Drai
- Laboratoire de Biochimie, Centre Hospitalier Lyon-Sud, Pierre Bénite, France; INSERM 1060 CARMEN CENS Université de Lyon, F-69622 Lyon, France
| | - Charlotte Cuerq
- Laboratoire de Biochimie, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| | - Sandrine Lemoine
- Service d'Explorations Fonctionnelles Rénales et Métaboliques, Hôpital Edouard Herriot, Lyon, France; INSERM 1060 CARMEN CENS Université de Lyon, F-69622 Lyon, France
| | - Aoumeur Hadj-Aissa
- Service d'Explorations Fonctionnelles Rénales et Métaboliques, Hôpital Edouard Herriot, Lyon, France
| | - Maurice Laville
- Département de Néphrologie-Dialyse-Nutrition, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre Bénite, France; INSERM 1060 CARMEN CENS Université de Lyon, F-69622 Lyon, France
| | - Denis Fouque
- Département de Néphrologie-Dialyse-Nutrition, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre Bénite, France; INSERM 1060 CARMEN CENS Université de Lyon, F-69622 Lyon, France.
| |
Collapse
|
84
|
Chen CD, Tung TY, Liang J, Zeldich E, Tucker Zhou TB, Turk BE, Abraham CR. Identification of cleavage sites leading to the shed form of the anti-aging protein klotho. Biochemistry 2014; 53:5579-87. [PMID: 25110992 PMCID: PMC4151695 DOI: 10.1021/bi500409n] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
![]()
Membrane
protein shedding is a critical step in many normal and
pathological processes. The anti-aging protein klotho (KL), mainly
expressed in kidney and brain, is secreted into the serum and CSF,
respectively. KL is proteolytically released, or shed, from the cell
surface by ADAM10 and ADAM17, which are the α-secretases that
also cleave the amyloid precursor protein and other proteins. The
transmembrane KL is a coreceptor with the FGF receptor for FGF23,
whereas the shed form acts as a circulating hormone. However, the
precise cleavage sites in KL are unknown. KL contains two major cleavage
sites: one close to the juxtamembrane region and another between the
KL1 and KL2 domains. We identified the cleavage site involved in KL
release by mutating potential sheddase(s) recognition sequences and
examining the production of the KL extracellular fragments in transfected
COS-7 cells. Deletion of amino acids T958 and L959 results in a 50–60%
reduction in KL shedding, and an additional P954E mutation results
in further reduction of KL shedding by 70–80%. Deletion of
amino acids 954–962 resulted in a 94% reduction in KL shedding.
This mutant also had moderately decreased cell surface expression,
yet had overall similar subcellular localization as that of WT KL,
as demonstrated by immunofluorescence. Cleavage-resistant mutants
could function as a FGFR coreceptor for FGF23, but they lost activity
as a soluble form of KL in proliferation and transcriptional reporter
assays. Cleavage between the KL1 and KL2 domains is dependent on juxtamembrane
cleavage. Our results shed light onto mechanisms underlying KL release
from the cell membrane and provide a target for potential pharmacologic
interventions aimed at regulating KL secretion.
Collapse
Affiliation(s)
- Ci-Di Chen
- Departments of Biochemistry, ‡Biology, and §Pharmacology and Experimental Therapeutics, Boston University School of Medicine , Boston, Massachusetts 02118, United States
| | | | | | | | | | | | | |
Collapse
|
85
|
Lee EY, Kim SS, Lee JS, Kim IJ, Song SH, Cha SK, Park KS, Kang JS, Chung CH. Soluble α-klotho as a novel biomarker in the early stage of nephropathy in patients with type 2 diabetes. PLoS One 2014; 9:e102984. [PMID: 25084095 PMCID: PMC4118846 DOI: 10.1371/journal.pone.0102984] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 06/24/2014] [Indexed: 01/25/2023] Open
Abstract
Objective Although α-klotho is known as an anti-aging, antioxidant, and cardio-renal protective protein, the clinical implications of soluble α-klotho levels in patients with diabetes have not been evaluated. Therefore, this study evaluated whether plasma and urinary α-klotho levels are associated with albuminuria in kidney disease in diabetes. Research Design and Methods A total of 147 patients with type 2 diabetes and 25 healthy control subjects were enrolled. The plasma and urine concentrations of α-klotho were analyzed by enzyme-linked immunosorbent assay. Results Plasma α-klotho (572.4 pg/mL [95% CI, 541.9–604.6 pg/mL] vs. 476.9 pg/mL [95% CI, 416.9–545.5 pg/mL]) and urinary α-klotho levels (59.8 pg/mg creatinine [95% CI, 43.6–82.0 pg/mg creatinine] vs. 21.0 pg/mg creatinine [95% CI, 9.7–45.6 pg/mg creatinine]) were significantly higher in diabetic patients than non-diabetic controls. Among diabetic patients, plasma α-klotho concentration was inversely associated with albuminuria stages (normoalbuminuria, 612.6 pg/mL [95% CI, 568.9–659.6 pg/mL], microalbuminuria, 551.8 pg/mL [95% CI, 500.5–608.3 pg/mL], and macroalbuminuria, 505.7 pg/mL [95% CI, 439.7–581.7 pg/mL] (p for trend = 0.0081), while urinary α-klotho levels were remained constantly high with increasing urinary albumin excretion. Conclusions Soluble α-klotho levels in plasma and urine may be novel and useful early markers of diabetic renal injury.
Collapse
Affiliation(s)
- Eun Young Lee
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
- * E-mail:
| | - Sang Soo Kim
- Department of Internal Medicine, Pusan National University Hospital, Busan, Korea
| | - Ji-Sung Lee
- Biostatistical Consulting Unit, Soonchunhyang University Medical Center, Seoul, Korea
| | - In Joo Kim
- Department of Internal Medicine, Pusan National University Hospital, Busan, Korea
| | - Sang Heon Song
- Department of Internal Medicine, Pusan National University Hospital, Busan, Korea
| | - Seung-Kuy Cha
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Kyu-Sang Park
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Jeong Suk Kang
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Choon Hee Chung
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
86
|
Donate-Correa J, Muros de Fuentes M, Mora-Fernández C, Navarro-González JF. Pathophysiological implications of fibroblast growth factor-23 and Klotho and their potential role as clinical biomarkers. Clin Chem 2014; 60:933-940. [PMID: 23757358 DOI: 10.1373/clinchem.2013.206649] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Fibroblast growth factor-23 (FGF-23) and Klotho constitute the main regulatory system of phosphorus homeostasis. Beyond this physiological role, there is growing evidence suggesting that this system has relevant pathophysiological implications in different clinical processes. CONTENT In this review we discuss the pathophysiological implications of the FGF-23/Klotho system and the potential utility that measurements of its components may have as clinical biomarkers in different clinical settings, such as progression of chronic kidney disease, acute renal failure, and secondary hyperparathyroidism, as well as vascular dysfunction, atherosclerosis, and cardiovascular morbidity and mortality. We outline and discuss the current commercially available assays for determination of FGF-23 and Klotho and the assay limitations that must be overcome to translate these biomarkers into reliable indicators in clinical practice. SUMMARY In addition to its physiological role, the FGF-23/Klotho system appears to provide important information regarding the pathophysiology of several clinical conditions. Although there has been increasing study of the components of this new biological system and their potential use as clinical biomarkers, the ultimate value of this system in clinical practice will not be known until remaining assay limitations can be overcome and adequately designed studies have been conducted to demonstrate its clinical utility.
Collapse
Affiliation(s)
- Javier Donate-Correa
- Research Unit, GEENDIAB and REDINREN (Sociedad Española de Nefrología e Instituto de Salud Carlos III, RD12/0021/0019), Madrid, Spain
| | - Mercedes Muros de Fuentes
- Clinical Analysis Service, and GEENDIAB and REDINREN (Sociedad Española de Nefrología e Instituto de Salud Carlos III, RD12/0021/0019), Madrid, Spain
| | - Carmen Mora-Fernández
- Research Unit, GEENDIAB and REDINREN (Sociedad Española de Nefrología e Instituto de Salud Carlos III, RD12/0021/0019), Madrid, Spain
| | - Juan F Navarro-González
- Research Unit, Nephrology Service, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain; GEENDIAB and REDINREN (Sociedad Española de Nefrología e Instituto de Salud Carlos III, RD12/0021/0019), Madrid, Spain.
| |
Collapse
|
87
|
Assessment of tubular reabsorption of phosphate as a surrogate marker for phosphate regulation in chronic kidney disease. Clin Exp Nephrol 2014; 19:208-15. [PMID: 24682550 DOI: 10.1007/s10157-014-0962-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 03/07/2014] [Indexed: 12/17/2022]
Abstract
BACKGROUND/AIMS Fibroblast growth factor 23 (FGF23) and soluble α-Klotho are emerging potential biomarkers of phosphorus and vitamin D metabolism which change in concentration in early chronic kidney disease (CKD) in order to maintain normal phosphorus levels. Tubular reabsorption of phosphate (TRP) has been commonly used to assess renal tubular phosphate transport. The aim of this study was to evaluate the usefulness of TRP as a surrogate marker of parameters of CKD-mineral bone disease (CKD-MBD) in CKD. METHODS A cross-sectional study was performed in 93 stable patients with predialysis CKD stage 1-5. In all patients, TRP, estimated glomerular filtration rate (eGFR), calcium, phosphate, intact parathyroid hormone (iPTH), 25-hydroxyvitamin D, serum FGF23 and urine soluble α-Klotho levels were measured. RESULTS As renal function declined, TRP significantly decreased (P < 0.001; r = 0.763) and both iPTH and serum FGF23 increased (P < 0.001; r = -0.598, P < 0.001; r = -0.453, respectively). The prevalence of hyperphosphatemia, secondary hyperparathyroidism, FGF23 excess and abnormal TRP increased progressively with declining eGFR. Although TRP level changed later than FGF23, abnormal levels of both TRP and FGF23 were observed earlier than changes in iPTH and serum phosphate. Decreased TRP was found to be independently associated with decreased eGFR and increased iPTH but was not associated with urine soluble α-Klotho or serum FGF23 level in multiple linear regression analysis. CONCLUSION TRP is a simple, useful and cost-saving surrogate marker of the assessment of altered mineral metabolism in CKD patients and can be used as an alternative to serum FGF23, especially for mild to moderate renal insufficiency.
Collapse
|
88
|
Abstract
PURPOSE OF REVIEW The wealth of data regarding fibroblast growth factor-23 (FGF23) and Klotho in chronic kidney disease (CKD) has risen exponentially over the past decade. This review is an attempt to summarize pivotal aspects of previous research, provide an update of recent findings and define important areas for future investigation. RECENT FINDINGS The phosphaturic hormone FGF23 increases dramatically as renal function declines. Identification of contributing stimuli to the rise in FGF23 is fundamental and recent evidence suggest a multifactorial cause which entails perturbed osteocyte function and renal mechanisms such as Klotho deficiency and, somewhat paradoxically, systemic Klotho excess. Circulating FGF23 predicts adverse outcomes, particularly cardiovascular disease, in CKD as well as in the general population. The concept of FGF23 merely as a biomarker and regulator of mineral metabolism is currently challenged by data linking FGF23 to pathological processes such as cardiac hypertrophy. Conversely, tissue level of the FGF23 coreceptor Klotho declines in early CKD and this deficiency is linked to accelerated ageing, cellular senescence, vascular calcification, oxidative stress and renal fibrosis. At present, methodological difficulties limit the utility of soluble Klotho measurements. Animal proof-of-concept studies have demonstrated beneficial effects of Klotho delivery in CKD, whereas anti-FGF23 therapy using neutralizing antibodies improved biochemical and bone parameters at the expense of increased vascular calcification and mortality. SUMMARY Pathological alterations of FGF23-Klotho in CKD are implicated as clinical biomarkers and may provide novel therapeutic strategies to alleviate the cardiovascular risk and slow CKD progression.
Collapse
|
89
|
Chen J, Zhang X, Zhang H, Lin J, Zhang C, Wu Q, Ding X. Elevated Klotho promoter methylation is associated with severity of chronic kidney disease. PLoS One 2013; 8:e79856. [PMID: 24224012 PMCID: PMC3818221 DOI: 10.1371/journal.pone.0079856] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 09/25/2013] [Indexed: 12/14/2022] Open
Abstract
Klotho (KL) expression is down-regulated in the renal tissues of chronic kidney disease (CKD) animal models and patients with end-stage renal disease. The putative role of KL promoter hypermethylation in the progression of CKD remains unclear. The present study aimed to determine renal and peripheral blood mononuclear cells (PBMC) levels of KL promoter methylation and analyze their relationship with clinical and histological severity in patients with CKD. Using bisulfite pyrosequencing, renal and PBMC levels of KL promoter methylation were quantified in 47 patients with CKD. 47 nephrectomy specimens of patients with renal cell carcinoma and 48 PBMC specimens of healthy volunteers were used as renal tissue and PBMC controls, respectively. Renal expression of KL protein was assayed by immunohistochemistry staining. Receiver operating characteristic (ROC) curve was used to identify the optimal cut-off value of PBMC KL promoter methylation level for renal KL promoter hypermethylation. Higher levels of KL promoter methylation were observed in renal tissue and PBMC in patients with CKD compared with controls (8.79±3.24 vs. 5.17±1.11%, P<0.001; 7.20±2.79 vs. 3.27±0.79%, P<0.001). In these patients, renal KL methylation level correlated inversely with renal KL immunostaining intensity (ρ=-0.794, P<0.001). Estimated glomerular filtration rate correlated inversely with renal and PBMC levels of KL promoter methylation (r=-0.829, P<0.001; r=-0.645, P<0.001), while tubulointerstistial fibrosis score correlated positively (ρ=0.826, P<0.001; ρ=0.755, P<0.001). PBMC KL promoter methylation level correlated positively with renal KL promoter methylation level in patients with CKD (r=0.787, P<0.001). In ROC curve, the area under curve was 0.964 (P<0.001) and the optimal cut-off value was 5.83% with a sensitivity of 93.8% and specificity of 86.7% to predict renal KL promoter hypermethylation. The degree of KL promoter methylation is associated with clinical and histological severity of CKD. PBMC KL promoter methylation level may act as a potential biomarker of renal KL promoter hypermethylation.
Collapse
Affiliation(s)
- Jing Chen
- Laboratory of Kidney Disease, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoyan Zhang
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- * E-mail: (XD); (XZ)
| | - Han Zhang
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing Lin
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen Zhang
- School of Public Health, Fudan University, Shanghai, China
| | - Qing Wu
- School of Public Health, Fudan University, Shanghai, China
| | - Xiaoqiang Ding
- Laboratory of Kidney Disease, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Nephrology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- * E-mail: (XD); (XZ)
| |
Collapse
|
90
|
Abstract
Acromegaly is characterized by excessively high GH and IGF1 levels. Recent data suggest that soluble Klotho (sKlotho) is also elevated in patients with active acromegaly. sKlotho decreases towards normal following removal of the GH-producing pituitary adenoma. The Klotho gene was identified in mice following its accidental disruption by ectopic DNA. It is an ageing suppressor gene of restricted expression (mainly in kidneys, brain, and parathyroid and pituitary glands) encoding a transmembrane protein, mKlotho. mKlotho serves as a co-receptor in fibroblast growth factor 23 (FGF23) signalling. FGF23 promotes urinary phosphate excretion and inhibits the synthesis of calcitriol. The ectodomain of mKlotho is enzymatically released to result in a humoral factor, sKlotho, which exerts systemic effects (on ion channels and signalling pathways), possibly by working as an enzyme that modifies glycans of cell surface glycoproteins. GH enhances renal phosphate reabsorption and calcitriol production, i.e. exerts effects in the proximal tubule opposing those attributed to mKlotho, and attenuates calciuria in the distal tubule similar to sKlotho. sKlotho can be measured in extracellular fluids (serum, urine and cerebrospinal fluid (CSF)) by an ELISA. In line with predominant expression of Klotho in kidneys and choroid plexus, concentrations of sKlotho are particularly high in urine and CSF. Determination of sKlotho in serum and urine (both presumably reflecting GH action on the kidneys) could be used as a supplementary tool in the diagnosis and follow-up of patients with acromegaly. The question arises whether GH exerts selected actions via modifying activities of Klotho.
Collapse
Affiliation(s)
- Christoph Schmid
- Division of Endocrinology and Diabetology, University Hospital Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland Department of Neurosurgery, University Hospital Zurich, Frauenklinikstrasse 10, 8091 Zurich, Switzerland Division of Endocrinology and Diabetology, Kantonsspital St Gallen, 9007 St Gallen, Switzerland Department of Neurosurgery, Clinic Hirslanden, Witellikerstrasse 40, 8032 Zurich, Switzerland
| | | | | | | | | |
Collapse
|
91
|
Craver L, Dusso A, Martinez-Alonso M, Sarro F, Valdivielso JM, Fernández E. A low fractional excretion of Phosphate/Fgf23 ratio is associated with severe abdominal Aortic calcification in stage 3 and 4 kidney disease patients. BMC Nephrol 2013; 14:221. [PMID: 24119158 PMCID: PMC3852798 DOI: 10.1186/1471-2369-14-221] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 10/09/2013] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Vascular calcification (VC) contributes to high mortality rates in chronic kidney disease (CKD). High serum phosphate and FGF23 levels and impaired phosphaturic response to FGF23 may affect VC. Therefore, their relative contribution to abdominal aortic calcification (AAC) was examined in patients CKD stages 3-4. METHODS Potential risk factors for AAC, measured by the Kauppila Index (KI), were studied in 178 patients. RESULTS In multivariate linear analysis, AAC associated positively with age, male gender, CKD-stage, presence of carotid plaques (CP) and also with FGF23, but negatively with fractional excretion of phosphate (FEP). Intriguingly, FEP increased with similar slopes with elevations in PTH, with reductions in GFR, and also with elevations in FGF23 but the latter only in patients with none (KI = 0) or mild (KI = 1-5) AAC. Lack of a FEP-FGF23 correlation in patients with severe AAC (KI > 5) suggested a role for an impaired phosphaturic response to FGF23 but not to PTH in AAC. Logistic and zero-inflated analysis confirmed the independent association of age, CKD stage, male gender and CP with AAC, and also identified a threshold FEP/FGF23 ratio of 1/3.9, below which the chances for a patient of presenting severe AAC increased by 3-fold. Accordingly, KI remained unchanged as FEP/FGF23 ratios decreased from 1/1 to 1/3.9 but markedly increased in parallel with further reductions in FEP/FGF23 < 1/3.9. CONCLUSIONS In CKD 3-4, an impaired phosphaturic response to FGF23 with FEP/FGF23 < 1/3.9 associates with severe AAC independently of age, gender or CP.
Collapse
Affiliation(s)
- Lourdes Craver
- Nephrology Service and Unit for the Detection and Treatment of Atherothrombotic diseases (UDETMA), Hospital Universitari Arnau de Vilanova, Av Rovira Roure, 25198 Lleida, Spain.
| | | | | | | | | | | |
Collapse
|
92
|
Miranda J, Romero R, Korzeniewski SJ, Schwartz AG, Chaemsaithong P, Stampalija T, Yeo L, Dong Z, Hassan SS, Chrousos GP, Gold P, Chaiworapongsa T. The anti-aging factor α-klotho during human pregnancy and its expression in pregnancies complicated by small-for-gestational-age neonates and/or preeclampsia. J Matern Fetal Neonatal Med 2013; 27:449-57. [PMID: 23808483 DOI: 10.3109/14767058.2013.818652] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE α-klotho, a protein with anti-aging properties, has been involved in important biological processes, such as calcium/phosphate metabolism, resistance to oxidative stress, and nitric oxide production in the endothelium. Recent studies have suggested a role of α-klotho in endocrine regulation of mineral metabolism and postnatal growth in infants. Yet, the role of α-klotho during pregnancy remains largely unknown. The aim of this study was to determine whether maternal plasma concentration of α-klotho changes during pregnancy and evaluate its expression in pregnancies complicated by small for gestational age (SGA) and/or preeclampsia (PE). STUDY DESIGN This cross-sectional study included patients in the following groups: (1) non pregnant women (n = 37); (2) uncomplicated pregnancy (n = 130); (3) PE without an SGA neonate (PE; n = 58); (4) PE with an SGA neonate (PE and SGA; n = 52); and (5) SGA neonate without PE (SGA; n = 52). Plasma concentrations of α-klotho were determined by ELISA. RESULTS The median plasma α-klotho concentration was higher in pregnant than in non-pregnant women. Among women with an uncomplicated pregnancy, the median plasma concentration of α-klotho increased as a function of gestational age (Spearman Rho = 0.2; p = 0.006). The median (interquartile range) plasma concentration of α-klotho in women with PE and SGA [947.6 (762-2013) pg/mL] and SGA without PE [1000 (585-1567) pg/mL] were 21% and 17% lower than that observed in women with an uncomplicated pregnancy [1206.6 (894-2012) pg/mL], (p = 0.005 and p = 0.02), respectively. Additionally, there were no significant differences in the median plasma concentration of α-klotho between uncomplicated pregnancies and women with PE without an SGA neonate (p = 0.5). CONCLUSION Maternal plasma concentration of α-klotho was higher during pregnancy than in a non-pregnant state. Moreover, the median maternal plasma concentration of α-klotho was lower in mothers who delivered an SGA neonate than in those with an uncomplicated pregnancy regardless of the presence or absence of PE.
Collapse
Affiliation(s)
- Jezid Miranda
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda , MD and Detroit, MI , USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Karalliedde J, Maltese G, Hill B, Viberti G, Gnudi L. Effect of renin-angiotensin system blockade on soluble Klotho in patients with type 2 diabetes, systolic hypertension, and albuminuria. Clin J Am Soc Nephrol 2013; 8:1899-905. [PMID: 23929932 DOI: 10.2215/cjn.02700313] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND OBJECTIVES Soluble Klotho is an anti-aging phosphaturic protein associated with vascular-renal protection. In vitro and in vivo studies have demonstrated that renin-angiotensin system (RAS) blockade increases soluble Klotho levels. The effect of RAS blockers on soluble Klotho in patients with diabetic kidney disease (DKD) is unknown. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Plasma-soluble Klotho was measured in a secondary analysis of a randomized controlled clinical trial performed at a single university hospital center (ClinicalTrials.gov number NCT001715, from March 2003 to September 2006). Seventy-six patients with type 2 diabetes and DKD (all with albuminuria and serum creatinine <1.7 mg/dl) were studied at baseline and at 24 weeks (study end) after randomization to valsartan/hydrochlorothiazide (n=37) or amlodipine (n=39) treatment. Aortic-pulse wave velocity by applanation tonometry and albuminuria (from three timed urine collections) were also measured at baseline and 24 weeks. RESULTS Valsartan/hydrochlorothiazide treatment significantly increased mean (± SD) soluble Klotho (from 432.7 ± 179 to 506.4 ± 226.8 pg/ml; P=0.01) and reduced serum phosphate (from 3.25 ± 1.18 to 2.60 ± 0.96 mg/dl; P=0.04) compared with amlodipine (from 430.1 ± 145.8 to 411.9 ± 157.6 pg/ml and from 2.94 ± 0.56 to 2.69 ± 1.52 mg/dl, respectively). There was a significant difference between treatment groups in soluble Klotho (mean 91.9 pg/ml; 95% confidence interval, 19.9 to 162) and serum phosphate levels (mean -0.68 mg/dl; 95% confidence interval, -0.15 to -1.33) with valsartan/hydrochlorothiazide treatment (P=0.03 and P=0.04, respectively). Attained BP was similar in the two groups and levels of soluble Klotho were not associated with aortic-pulse wave velocity and albuminuria, variables that fell significantly only with valsartan/hydrochlorothiazide. CONCLUSIONS Treatment with a RAS blocker, valsartan, is associated with an increase in soluble Klotho, which may contribute to the BP-independent cardiorenal benefits of these drugs in DKD.
Collapse
|
94
|
Kohler S, Tschopp O, Sze L, Neidert M, Bernays RL, Spanaus KS, Wiesli P, Schmid C. Monitoring for potential residual disease activity by serum insulin-like growth factor 1 and soluble Klotho in patients with acromegaly after pituitary surgery: is there an impact of the genomic deletion of exon 3 in the growth hormone receptor (d3-GHR) gene on "safe" GH cut-off values? Gen Comp Endocrinol 2013; 188:282-7. [PMID: 23648743 DOI: 10.1016/j.ygcen.2013.04.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 04/16/2013] [Accepted: 04/19/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND Acromegaly is an illness usually defined by excessively high growth hormone (GH) and insulin like growth factor 1 (IGF-1) levels, the latter mainly reflecting GH action on the liver. IGF-1, also known as somatomedin C, mediates several actions of GH. The diagnosis and management of acromegaly is relatively straight forward, but long-term follow-up of patients can be difficult, as elevated IGF-1 levels can occur in the presence of apparently normalised GH levels and late recurrence of acromegaly may arise despite previous suppression on oral glucose tolerance testing. Data suggest this applies especially to patients in whom the GH receptor lacks exon 3. In such patients, GH may not always be a useful marker of disease, and traditional GH cut-offs may be misleading. Recent data suggest that soluble Klotho (sKlotho), besides and in addition to IGF-1, may help monitor the activity of GH-producing adenomas (presumably reflecting GH action on the kidneys) and may be a useful supplementary tool. METHODS GHR genotyping was performed in 112 patients with acromegaly. IGF-1 and sKlotho levels were measured in the sera of patients before and after transsphenoidal surgery, with emphasis on patients judged inconclusively cured by surgery or with small residual tumour masses shortly after surgery. Patients were assessed for recurrence of acromegaly with GH levels (random or nadir during an oGTT). RESULTS Of the 48 patients who underwent surgery between 2000 and 2009 and who had well-documented longer term follow-up at our institution, 29 had no biochemical evidence of residual disease activity after transsphenoidal surgery (marked reduction in IGF-1 and sKlotho levels, GH suppressible to <1 ng/ml) and were classified as in remission. 2 of these patients developed recurrent symptoms of acromegaly during follow-up with increasing levels of IGF-1 and sKlotho, and both patients were carriers of the d3-GHR genotype. CONCLUSIONS Acromegalic patients with the d3-GHR polymorphism might be - for a given low postsurgical GH level - at higher risk for recurrence and may require a lower GH nadir during oGTT to be classified as in remission. Soluble Klotho could be useful in the follow-up of acromegalic patients. The question arises whether sKlotho not only reflects the activity of GH-secreting pituitary adenomas but whether Klotho (ectodomain clipping?) could also mediate selected actions of GH.
Collapse
Affiliation(s)
- S Kohler
- Department of Internal Medicine, Division of Endocrinology and Diabetes, University Hospital Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
95
|
Heijboer AC, Blankenstein MA, Hoenderop J, de Borst MH, Vervloet MG. Laboratory aspects of circulating α-Klotho. Nephrol Dial Transplant 2013; 28:2283-7. [PMID: 23787542 DOI: 10.1093/ndt/gft236] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND α-Klotho is a protein mainly produced in the kidney. Its circulating form has been suggested to link renal damage and distant tissue pathology. As three assays to measure α-Klotho became commercially available, we performed an evaluation of these commercially available Klotho assays. METHODS We studied within-run variation, between-run variation, matrix effects, linearity, and recovery of added recombinant human Klotho in the α-Klotho assays of IBL (IBL International GmbH, Hamburg, Germany), Cusabio (Cusabio Biotech, Wuhan, China) and USCN (USCN life Science, Inc., Wuhan, China) using both serum and ethylenediaminetetraacetic acid plasma. RESULTS Within run variation was 4, 13 and 32% for the IBL, Cusabio and USCN assay, respectively. Agreement between serum and EDTA plasma was good in the IBL assay, but poor in the USCN and Cusabio assays however improved after modifications in the Cusabio assay. Standardization and agreement between assays was poor. CONCLUSIONS The commercially available methods for the measurement of α-Klotho differ in quality. Some of the manufacturers should improve their assays in order to produce accurate results so that reliable conclusions can be drawn from studies in which these assays are used.
Collapse
Affiliation(s)
- Annemieke C Heijboer
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, the Netherlands
| | | | | | | | | | | |
Collapse
|