1
|
Pannu CD, Baxter D, Anwar H. Spinal phosphaturic mesenchymal tumors: a rare etiology causing tumor-induced osteomalacia-a review of experience at a UK tertiary referral center and literature review. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2024; 33:1911-1920. [PMID: 37964171 DOI: 10.1007/s00586-023-08031-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/21/2023] [Accepted: 10/25/2023] [Indexed: 11/16/2023]
Abstract
PURPOSE This article aims to provide a comprehensive review of the management challenges associated with Spinal Phosphaturic Mesenchymal tumors (PMTs) and evaluates the surgical management outcomes for this rare entity linked to Tumor-induced osteolysis. The primary objective of this study is to enhance the familiarity of treating physicians with the clinical features, diagnosis, and treatment options for Spinal PMTs. METHODS A retrospective analysis was conducted, reviewing electronic medical records of patients diagnosed with spinal PMTs at our hospital between January 2019 and December 2022. The data collected included demographic information, clinical presentation, radiological findings, surgical details, and follow-up outcomes. RESULTS A total of three cases of Spinal PMTs causing Tumor-induced osteomalacia were identified. The diagnosis of Spinal PMTs presented challenges, with incidental detection often occurring during routine imaging. Surgical management was undertaken, resulting in successful symptom resolution and normalization of phosphate levels. The application of 68 Ga-DOTA-TATE PET/CT scans facilitated tumor localization, aiding in surgical planning. Spinal PMTs demonstrated a favorable response to surgical intervention. CONCLUSION Spinal PMTs play a significant role in Tumor-induced osteolysis, warranting timely and accurate diagnosis. Although diagnosing Spinal PMTs presents challenges, surgical management has proven to yield favorable outcomes, effectively alleviating symptoms and restoring phosphate levels. A multidisciplinary approach and continued vigilance are essential in ensuring early diagnosis, effective treatment, and long-term monitoring for patients affected by spinal PMTs.
Collapse
Affiliation(s)
- Chaitanya Dev Pannu
- Royal National Orthopedic Hospital, Brockley Hill, Stanmore, London, HA7 4LP, UK.
| | - David Baxter
- Royal National Orthopedic Hospital, Brockley Hill, Stanmore, London, HA7 4LP, UK
| | - Hanny Anwar
- Royal National Orthopedic Hospital, Brockley Hill, Stanmore, London, HA7 4LP, UK
| |
Collapse
|
2
|
Xiong A, Zhou S, Liu C, Hu Z, Xie C, Zheng X, Luo X. Vitamin D levels in idiopathic inflammatory myopathy patients: a meta-analysis. Postgrad Med 2024; 136:141-149. [PMID: 38420733 DOI: 10.1080/00325481.2024.2325335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/26/2024] [Indexed: 03/02/2024]
Abstract
PURPOSE This meta-analysis aimed to explore correlations between vitamin D and idiopathic inflammatory myopathy (IIM). METHODS A comprehensive database search was conducted on 13 October 2020. Mean differences (MDs) and aggregated risk ratios (RR) with 95% confidence intervals (CIs) were used to determine the correlation between vitamin D deficiency (VDD) and IIM. Statistical analysis was performed with RevMan 5.4 and Stata15, statistical significance was set at p < 0.05. RESULTS Search revealed five studies with 286 IIM patients and 480 healthy controls. Results with random-effects modeling indicated that serum vitamin D levels were significantly lower in IIM patients than in healthy controls (MD = -13.10 ng/mL; 95% CI: -16.51 to -9.68; p < 0.00001). No differences were found between patients with IIM and other autoimmune diseases on vitamin D levels (MD =-2.65 ng/mL; 95% CI: -11.31-6.01; p = 0.55). In two studies with 185 IIM patients, those with low vitamin D levels exhibited higher creatine kinase levels (MD = 85.20 IU/L; 95% CI: 72.67-97.73; p < 0.00001) than those with normal vitamin D levels. VDD was correlated with an increased risk of IIM (RR = 3.24, 95% CI: 1.81-5.79; p < 0.0001). CONCLUSION This meta-analysis showed correlations between vitamin D level and IIM. The results indicated, VDD may be a risk factor for IIM, a determinant of immune dysregulation in IIM, or a consequence of IIM. Also, it implied further research to determine whether vitamin D supplementation is beneficial for patients with IIM.
Collapse
Affiliation(s)
- Anji Xiong
- Department of Rheumatology and Immunology, Nanchong Central Hospital, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong Hospital of Beijing Anzhen Hospital Capital Medical University, Nanchong, China
- Inflammation and Immunology Key Laboratory of Nanchong City, Nanchong, Sichuan, China
- Nanchong Central Hospital (Nanchong Clinical Research Center), Nanchong, China
| | - Shifeng Zhou
- Department of Rheumatology and Immunology, Nanchong Central Hospital, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong Hospital of Beijing Anzhen Hospital Capital Medical University, Nanchong, China
| | - Can Liu
- Department of Rheumatology and Immunology, Nanchong Central Hospital, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong Hospital of Beijing Anzhen Hospital Capital Medical University, Nanchong, China
| | - Ziyi Hu
- Department of Rheumatology and Immunology, Nanchong Central Hospital, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong Hospital of Beijing Anzhen Hospital Capital Medical University, Nanchong, China
| | - Chuanmei Xie
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiaoli Zheng
- School of Basic Medicine, Southwest Medical University
| | - Xiongyan Luo
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University
| |
Collapse
|
3
|
Sarathi V, Dhananjaya MS, Karlekar M, Lila AR. Vitamin D deficiency or resistance and hypophosphatemia. Best Pract Res Clin Endocrinol Metab 2024; 38:101876. [PMID: 38365463 DOI: 10.1016/j.beem.2024.101876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Abstract
Vitamin D is mainly produced in the skin (cholecalciferol) by sun exposure while a fraction of it is obtained from dietary sources (ergocalciferol). Vitamin D is further processed to 25-hydroxyvitamin D and 1,25-dihydroxy vitamin D (calcitriol) in the liver and kidneys, respectively. Calcitriol is the active form which mediates the actions of vitamin D via vitamin D receptor (VDR) which is present ubiquitously. Defect at any level in this pathway leads to vitamin D deficient or resistant rickets. Nutritional vitamin D deficiency is the leading cause of rickets and osteomalacia worldwide and responds well to vitamin D supplementation. Inherited disorders of vitamin D metabolism (vitamin D-dependent rickets, VDDR) account for a small proportion of calcipenic rickets/osteomalacia. Defective 1α hydroxylation of vitamin D, 25 hydroxylation of vitamin D, and vitamin D receptor result in VDDR1A, VDDR1B and VDDR2A, respectively whereas defective binding of vitamin D to vitamin D response element due to overexpression of heterogeneous nuclear ribonucleoprotein and accelerated vitamin D metabolism cause VDDR2B and VDDR3, respectively. Impaired dietary calcium absorption and consequent calcium deficiency increases parathyroid hormone in these disorders resulting in phosphaturia and hypophosphatemia. Hypophosphatemia is a common feature of all these disorders, though not a sine-qua-non and leads to hypomineralisation of the bone and myopathy. Improvement in hypophosphatemia is one of the earliest markers of response to vitamin D supplementation in nutritional rickets/osteomalacia and the lack of such a response should prompt evaluation for inherited forms of rickets/osteomalacia.
Collapse
Affiliation(s)
- Vijaya Sarathi
- Department of Endocrinology, Vydehi Institute of Medical Sciences and Research Center, Bengaluru 560066, India.
| | | | - Manjiri Karlekar
- Department of Endocrinology, Seth G S Medical College and King Edward Hospital, Mumbai 400012, India
| | - Anurag Ranjan Lila
- Department of Endocrinology, Seth G S Medical College and King Edward Hospital, Mumbai 400012, India
| |
Collapse
|
4
|
Stawicki MK, Abramowicz P, Sokolowska G, Wołejszo S, Grant WB, Konstantynowicz J. Can vitamin D be an adjuvant therapy for juvenile rheumatic diseases? Rheumatol Int 2023; 43:1993-2009. [PMID: 37566255 PMCID: PMC10495493 DOI: 10.1007/s00296-023-05411-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023]
Abstract
Vitamin D, known for its essential role in calcium and bone homeostasis, has multiple effects beyond the skeleton, including regulation of immunity and modulation of autoimmune processes. Several reports have shown suboptimal serum 25 hydroxyvitamin D [25(OH)D] levels in people with different inflammatory and autoimmune rheumatic conditions, and an association between 25(OH)D levels, disease activity and outcomes. Although most available data pertain to adults, insights often are extended to children. Juvenile rheumatic diseases (JRDs) are a significant health problem during growth because of their complex pathogenesis, chronic nature, multisystemic involvement, and long-term consequences. So far, there is no definitive or clear evidence to confirm the preventive or therapeutic effect of vitamin D supplementation in JRDs, because results from randomized controlled trials (RCTs) have produced inconsistent outcomes. This review aims to explore and discuss the potential role of vitamin D in treating selected JRDs. Medline/PubMed, EMBASE, and Scopus were comprehensively searched in June 2023 for any study on vitamin D supplementary role in treating the most common JRDs. We used the following keywords: "vitamin D" combined with the terms "juvenile idiopathic arthritis", "juvenile systemic scleroderma", "juvenile systemic lupus erythematosus", "juvenile inflammatory myopathies", "Behcet disease", "periodic fever syndromes" and "juvenile rheumatic diseases". Observational studies have found that serum 25(OH)D concentrations are lower in juvenile idiopathic arthritis, juvenile systemic lupus erythematosus, juvenile systemic scleroderma, Behcet disease and proinflammatory cytokine concentrations are higher. This suggests that vitamin D supplementation might be beneficial, however, current data are insufficient to confirm definitively the complementary role of vitamin D in the treatment of JRDs. Considering the high prevalence of vitamin D deficiency worldwide, children and adolescents should be encouraged to supplement vitamin D according to current recommendations. More interventional studies, especially well-designed RCTs, assessing the dose-response effect and adjuvant effect in specific diseases, are needed to determine the potential significance of vitamin D in JRDs treatment.
Collapse
Affiliation(s)
- Maciej K. Stawicki
- Department of Pediatrics, Rheumatology, Immunology, and Metabolic Bone Diseases, Medical University of Bialystok, University Children’s Clinical Hospital in Bialystok, Waszyngtona Street 17, 15274 Bialystok, Poland
| | - Paweł Abramowicz
- Department of Pediatrics, Rheumatology, Immunology, and Metabolic Bone Diseases, Medical University of Bialystok, University Children’s Clinical Hospital in Bialystok, Waszyngtona Street 17, 15274 Bialystok, Poland
| | | | - Sebastian Wołejszo
- Department of Pediatrics, Rheumatology, Immunology, and Metabolic Bone Diseases, Medical University of Bialystok, University Children’s Clinical Hospital in Bialystok, Waszyngtona Street 17, 15274 Bialystok, Poland
| | - William B. Grant
- Sunlight, Nutrition, and Health Research Center, San Francisco, CA USA
| | - Jerzy Konstantynowicz
- Department of Pediatrics, Rheumatology, Immunology, and Metabolic Bone Diseases, Medical University of Bialystok, University Children’s Clinical Hospital in Bialystok, Waszyngtona Street 17, 15274 Bialystok, Poland
| |
Collapse
|
5
|
Donati S, Palmini G, Aurilia C, Falsetti I, Marini F, Giusti F, Iantomasi T, Brandi ML. Calcifediol: Mechanisms of Action. Nutrients 2023; 15:4409. [PMID: 37892484 PMCID: PMC10610216 DOI: 10.3390/nu15204409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Due to its essential role in calcium and phosphate homeostasis, the secosteroid hormone calcitriol has received growing attention over the last few years. Calcitriol, like other steroid hormones, may function through both genomic and non-genomic mechanisms. In the traditional function, the interaction between the biologically active form of vitamin D and the vitamin D receptor (VDR) affects the transcription of thousands of genes by binding to repeated sequences present in their promoter region, named vitamin D-responsive elements (VDREs). Non-transcriptional effects, on the other hand, occur quickly and are unaffected by inhibitors of transcription and protein synthesis. Recently, calcifediol, the immediate precursor metabolite of calcitriol, has also been shown to bind to the VDR with weaker affinity than calcitriol, thus exerting gene-regulatory properties. Moreover, calcifediol may also trigger rapid non-genomic responses through its interaction with specific membrane vitamin D receptors. Membrane-associated VDR (mVDR) and protein disulfide isomerase family A member 3 (Pdia3) are the best-studied candidates for mediating these rapid responses to vitamin D metabolites. This paper provides an overview of the calcifediol-related mechanisms of action, which may help to better understand the vitamin D endocrine system and to identify new therapeutic targets that could be important for treating diseases closely associated with vitamin D deficiency.
Collapse
Affiliation(s)
- Simone Donati
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (S.D.); (G.P.); (C.A.); (I.F.); (F.G.); (T.I.)
| | - Gaia Palmini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (S.D.); (G.P.); (C.A.); (I.F.); (F.G.); (T.I.)
| | - Cinzia Aurilia
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (S.D.); (G.P.); (C.A.); (I.F.); (F.G.); (T.I.)
| | - Irene Falsetti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (S.D.); (G.P.); (C.A.); (I.F.); (F.G.); (T.I.)
| | - Francesca Marini
- Fondazione Italiana Ricerca Sulle Malattie dell’Osso (FIRMO Onlus), 50129 Florence, Italy;
| | - Francesca Giusti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (S.D.); (G.P.); (C.A.); (I.F.); (F.G.); (T.I.)
| | - Teresa Iantomasi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (S.D.); (G.P.); (C.A.); (I.F.); (F.G.); (T.I.)
| | - Maria Luisa Brandi
- Fondazione Italiana Ricerca Sulle Malattie dell’Osso (FIRMO Onlus), 50129 Florence, Italy;
- Donatello Bone Clinic, Villa Donatello Hospital, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
6
|
董 沙, 车 若, 郑 必, 张 爱, 王 春, 白 咪, 陈 颖. [Value of serum fibroblast growth factor 23 in diagnosis of hypophosphatemic rickets in children]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:705-710. [PMID: 37529952 PMCID: PMC10414175 DOI: 10.7499/j.issn.1008-8830.2303016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/02/2023] [Indexed: 08/03/2023]
Abstract
OBJECTIVES To study the value of serum fibroblast growth factor 23 (FGF23) in the diagnosis of hypophosphatemic rickets in children. METHODS A total of 28 children who were diagnosed with hypophosphatemic rickets in Children's Hospital of Nanjing Medical University from January 2016 to June 2021 were included as the rickets group. Forty healthy children, matched for sex and age, who attended the Department of Child Healthcare of the hospital were included as the healthy control group. The serum level of FGF23 was compared between the two groups, and the correlations of the serum FGF23 level with clinical characteristics and laboratory test results were analyzed. The value of serum FGF23 in the diagnosis of hypophosphatemic rickets was assessed. RESULTS The rickets group had a significantly higher serum level of FGF23 than the healthy control group (P<0.05). In the rickets group, the serum FGF23 level was positively correlated with the serum alkaline phosphatase level (rs=0.38, P<0.05) and was negatively correlated with maximum renal tubular phosphorus uptake/glomerular filtration rate (rs=-0.64, P<0.05), while it was not correlated with age, height Z-score, sex, and parathyroid hormone (P>0.05). Serum FGF23 had a sensitivity of 0.821, a specificity of 0.925, an optimal cut-off value of 55.77 pg/mL, and an area under the curve of 0.874 in the diagnosis of hypophosphatemic rickets (P<0.05). CONCLUSIONS Serum FGF23 is of valuable in the diagnosis of hypophosphatemic rickets in children, which providing a theoretical basis for early diagnosis of this disease in clinical practice.
Collapse
Affiliation(s)
| | | | - 必霞 郑
- 南京医科大学附属儿童医院儿科学重点实验室,江苏南京210000
| | | | - 春莉 王
- 南京医科大学附属儿童医院儿科学重点实验室,江苏南京210000
| | - 咪 白
- 南京医科大学附属儿童医院儿科学重点实验室,江苏南京210000
| | | |
Collapse
|
7
|
Kubota M, Hamasaki Y, Hashimoto J, Aoki Y, Kawamura T, Saito A, Yuasa R, Muramatsu M, Komaba H, Toyoda M, Fukagawa M, Shishido S, Sakai K. Fibroblast growth factor 23-Klotho and mineral metabolism in the first year after pediatric kidney transplantation: A single-center prospective study. Pediatr Transplant 2023; 27:e14440. [PMID: 36471536 DOI: 10.1111/petr.14440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND The role of fibroblast growth factor 23 (FGF23) levels in mineral metabolism before and after kidney transplantation in pediatric patients is poorly understood. METHODS We prospectively evaluated 24 patients under 18 years of age (4.5 [3.3-9.8] years) who underwent living kidney transplantation between July 2016 and March 2018, and measured intact FGF23 and serum αKlotho levels, and other parameters of mineral metabolism before and after transplantation (Day 7, 1 and 4 months, and 1 year). Relationships between parameters were examined by linear analysis. RESULTS FGF23 level was 440.8 [63.4-5916.3] pg/ml pre-transplant and decreased significantly to 37.1 [16.0-71.5] pg/ml at Day 7 post-transplant (-91.6%, p < .001). Thereafter, it remained at normal levels until 1 year. αKlotho level was 785 [568-1292] pg/ml pre-transplant and remained low at Day 7 and 1 month post-transplant, with an increasing trend at 4 months. Post-transplant phosphorus levels were significantly decreased compared with pre-transplant, with a lowest level of 1.7 [1.3-2.9] mg/dl, -5.7 [-6.8, -3.8] SD at Day 4, followed by gradual recovery. Phosphorus levels and the ratio of tubular maximum phosphate reabsorption were significantly and negatively associated with pre-transplant FGF23 until 4 months of post-transplant. Pre-transplant αKlotho was negatively associated with pre-transplant FGF23 but not FGF23 or other parameters after transplantation. CONCLUSION FGF23 in pediatric kidney transplant patients decreased rapidly after transplantation and associated with post-transplant hypophosphatemia and increased phosphorus excretion. Post-transplant αKlotho was low early post-transplant but tended to increase subsequently. Post-transplant αKlotho was unaffected by pre-transplant FGF23 or other factors, suggesting pre-transplant chronic kidney disease status has no effect.
Collapse
Affiliation(s)
- Mai Kubota
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Yuko Hamasaki
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Junya Hashimoto
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Yujiro Aoki
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Takeshi Kawamura
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Akinobu Saito
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Rena Yuasa
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Masaki Muramatsu
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Hirotaka Komaba
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan
| | - Masao Toyoda
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan
| | - Masafumi Fukagawa
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan
| | - Seichiro Shishido
- Department of Pediatric Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| | - Ken Sakai
- Department of Nephrology, Toho University, Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Yimamu Y, Ohtani A, Takei Y, Furuichi A, Kamei Y, Yamanaka-Okumura H, Ohminami H, Masuda M, Miyazaki M, Yamamoto H, Taketani Y. 25-hydroxyvitamin D-1α-hydroxylase (CYP27B1) induces ectopic calcification. J Clin Biochem Nutr 2022; 71:103-111. [PMID: 36213783 PMCID: PMC9519415 DOI: 10.3164/jcbn.22-16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 02/15/2022] [Indexed: 12/04/2022] Open
Abstract
Vascular calcification is an important pathogenesis related to cardiovascular disease and high mortality rate in chronic kidney disease (CKD) patients. It has been well-known that hyper-phosphatemia induces osteochondrogenic transition of vascular smooth muscle cells (VSMCs) resulting ectopic calcification in aortic media, cardiac valve, and kidney. However, the detailed mechanism of the ectopic calcification has been not clarified yet. Here, we found that the co-localization of CYP27B1 with the calcified lesions of aorta and arteries in kidney of klotho mutant (kl/kl) mice, and then investigated the role of CYP27B1 in the mineralization of the VSMCs. Under high phosphate condition, overexpression of CYP27B1 induced calcification and osteocalcin mRNA expression in the VSMCs. Inversely, siRNA-CYP27B1 inhibited high phosphate-induced calcification of the VSMCs. We also found that the accumulated CYP27B1 protein was glycosylated in the kidney of kl/kl mice. Therefore, overexpression of CYP27B1-N310A and CYP27B1-T439A, which are a mutation for N-linked glycosylation site (N310A) and a mutation for O-linked glycosylation site (T439A) in CYP27B1, decreased calcium deposition and expression of RUNX2 induced by high phosphate medium in VSMCs compared with wild-type CYP27B1. These results suggest that extra-renal expression of glycosylated CYP27B1 would be required for ectopic calcification of VSMCs under hyperphosphatemia.
Collapse
Affiliation(s)
- Yilimulati Yimamu
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Ayako Ohtani
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yuichiro Takei
- Faculty of Nutrition, University of Kochi, 2751-1, Ike, Kochi 781-8515, Japan
| | - Airi Furuichi
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yuki Kamei
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hisami Yamanaka-Okumura
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hirokazu Ohminami
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Masashi Masuda
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Makoto Miyazaki
- Division of Renal Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, 13001 East 17th Place, Aurora, CO 80045, USA
| | - Hironori Yamamoto
- Department of Health and Nutrition, Faculty of Human Life, Jin-ai University, 3-1-1 Ohde-cho, Echizen, Fukui 915-8568, Japan
| | - Yutaka Taketani
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
9
|
Živanović J, Jarić I, Ajdžanović V, Miler M, Stanković S, Milošević V, Filipović B. Genistein regulates calcium and phosphate homeostasis without activation of MEK 1/2 signalling pathway in an animal model of the andropause. Ann Anat 2021; 239:151836. [PMID: 34563672 DOI: 10.1016/j.aanat.2021.151836] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 01/02/2023]
Abstract
Soy isoflavone genistein interplays with numerous physiological or pathophysiological processes during ageing. However, its protective role and underlying mechanisms of action in the regulation of calcium (Ca2+) and phosphate (Pi) homeostasis in an animal model of the andropause are yet to be fully clarified. Wistar male rats (16-month-old) were divided into sham-operated, orchidectomized, orchidectomized estradiol-treated (0.625 mg/kg b.m./day) and orchidectomized genistein-treated (30 mg/kg b.m./day) groups. Treatments were administered subcutaneously for 3 weeks, while the controls received vehicle alone. Estradiol treatment increased the expression level of fibroblast growth factor receptor (FGFR) and parathyroid hormone 1 receptor (PTH1R), and activated mitogen - activated protein kinase kinase 1/2 (MEK 1/2) signaling pathway in the kidneys. Genistein application induced a prominent gene and protein expression of Klotho and downregulated the expression of FGFR and PTH1R in the kidney of andropausal rats. Activation of protein kinase B (Akt) signalling pathway was observed, while MEK 1/2 signaling pathway wasn't altered after genistein treatment. The increase of 25 (OH) vitamin D in the serum and decrease in Ca2+ urine content was observed after genistein application. Our findings strongly suggest genistein as a potent biocompound with beneficial effects on the regulation of Ca2+ and Pi homeostasis, especially during aging process when the balance of mineral metabolism is impaired. These novel data provide closer insights into the physiological roles of genistein in the regulation of mineral homeostasis.
Collapse
Affiliation(s)
- Jasmina Živanović
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| | - Ivana Jarić
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia; Animal Welfare Division, Vetsuisse, University of Bern, Bern, Switzerland
| | - Vladimir Ajdžanović
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Marko Miler
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Sanja Stanković
- Center for Medical Biochemistry, Clinical Centre of Serbia, Belgrade, Serbia
| | - Verica Milošević
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Branko Filipović
- Department of Cytology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
10
|
Raikou VD. Serum phosphate and chronic kidney and cardiovascular disease: Phosphorus potential implications in general population. World J Nephrol 2021; 10:76-87. [PMID: 34631478 PMCID: PMC8477270 DOI: 10.5527/wjn.v10.i5.76] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/20/2021] [Accepted: 07/23/2021] [Indexed: 02/06/2023] Open
Abstract
It has already been established that in end-stage renal disease, hyperphosphatemia causes soft tissue calcification including vascular calcifications. It has also been supported that there is a connection between increased serum phosphate and morbidity in subjects, who suffer from renal disease. However, studies in these populations conferred mixed results. Several warnings are included in the role of serum phosphorus on cardiovascular disease in normal populations. Homeostasis of serum phosphate is obtained by the cooperation between regulatory hormones, cellular receptors and bone metabolic factors. There is the probability that one or more phosphate regulatory factors, rather than phosphate directly, may be responsible for observed associations with calcification and cardiovascular events in normal populations. Experimental studies have shown that the restriction of dietary phosphate prevents the progression of kidney dysfunction, although high dietary phosphate aggravates the renal function. In the current review, we discuss the role of serum phosphorus on progression of renal dysfunction and cardiovascular outcomes in chronic kidney disease patients and its involvement in important health risks in the general population.
Collapse
Affiliation(s)
- Vaia D Raikou
- Department of Nephrology, Doctors’ Hospital, Athens 11257, Greece
| |
Collapse
|
11
|
van der Vaart A, Yeung S, van Dijk P, Bakker S, de Borst M. Phosphate and fibroblast growth factor 23 in diabetes. Clin Sci (Lond) 2021; 135:1669-1687. [PMID: 34283205 PMCID: PMC8302806 DOI: 10.1042/cs20201290] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 12/11/2022]
Abstract
Diabetes is associated with a strongly elevated risk of cardiovascular disease, which is even more pronounced in patients with diabetic nephropathy. Currently available guideline-based efforts to correct traditional risk factors are only partly able to attenuate this risk, underlining the urge to identify novel treatment targets. Emerging data point towards a role for disturbances in phosphate metabolism in diabetes. In this review, we discuss the role of phosphate and the phosphate-regulating hormone fibroblast growth factor 23 (FGF23) in diabetes. We address deregulations of phosphate metabolism in patients with diabetes, including diabetic ketoacidosis. Moreover, we discuss potential adverse consequences of these deregulations, including the role of deregulated phosphate and glucose as drivers of vascular calcification propensity. Finally, we highlight potential treatment options to correct abnormalities in phosphate and FGF23. While further studies are needed to more precisely assess their clinical impact, deregulations in phosphate and FGF23 are promising potential target in diabetes and diabetic nephropathy.
Collapse
Affiliation(s)
- Amarens van der Vaart
- Department of Medicine, Division of Nephrology, University of Groningen, University Medical Centre Groningen, Groningen,The Netherlands
- Department of Medicine, Division of Endocrinology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Stanley M.H. Yeung
- Department of Medicine, Division of Nephrology, University of Groningen, University Medical Centre Groningen, Groningen,The Netherlands
| | - Peter R. van Dijk
- Department of Medicine, Division of Endocrinology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Stephan J.L. Bakker
- Department of Medicine, Division of Nephrology, University of Groningen, University Medical Centre Groningen, Groningen,The Netherlands
| | - Martin H. de Borst
- Department of Medicine, Division of Nephrology, University of Groningen, University Medical Centre Groningen, Groningen,The Netherlands
| |
Collapse
|
12
|
Mahajan K, Verma H, Choudhary S, Raju B, Silakari O. Identification of kinase inhibitors that rule out the CYP27B1-mediated activation of vitamin D: an integrated machine learning and structure-based drug designing approach. Mol Divers 2021; 25:1617-1641. [PMID: 34272637 DOI: 10.1007/s11030-021-10270-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 07/02/2021] [Indexed: 11/30/2022]
Abstract
CYP27B1, a cytochrome P450-containing hydroxylase enzyme, converts vitamin D precursor calcidiol (25-hydroxycholecalciferol) to its active form calcitriol (1α,25(OH)2D3). Tyrosine kinase inhibitor such as imatinib is reported to interfere with the activation of vitamin D3 by inhibiting CYP27B1 enzyme. Consequently, there is a decrease in the serum levels of active vitamin D that in turn may increase the relapse risk among the cancer patients treated with imatinib. Within this framework, the current study focuses on identifying other possible kinase inhibitors that may affect the calcitriol level in the body by inhibiting CYP27B1. To achieve this, we explored multiple machine learning approaches including support vector machine (SVM), random forest (RF), and artificial neural network (ANN) to identify possible CYP27B1 inhibitors from a pool of kinase inhibitors database. The most reliable classification model was obtained from the SVM approach with Matthews correlation coefficient of 0.82 for the external test set. This model was further employed for the virtual screening of kinase inhibitors from the binding database (DB), which tend to interfere with the CYP27B1-mediated activation of vitamin D. This screening yielded around 4646 kinase inhibitors that were further subjected to structure-based analyses using the homology model of CYP27B1, as the 3D structure of CYP27B1 complexed with heme was not available. Overall, five kinase inhibitors including two well-known drugs, i.e., AT7867 (Compound-2) and amitriptyline N-oxide (Compound-3), were found to interact with CYP27B1 in such a way that may preclude the conversion of vitamin D to its active form and hence testify the impairment of vitamin D activation pathway.
Collapse
Affiliation(s)
- Kanupriya Mahajan
- Molecular Modeling Lab, Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, Punjab, 147002, India
| | - Himanshu Verma
- Molecular Modeling Lab, Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, Punjab, 147002, India
| | - Shalki Choudhary
- Molecular Modeling Lab, Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, Punjab, 147002, India
| | - Baddipadige Raju
- Molecular Modeling Lab, Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, Punjab, 147002, India
| | - Om Silakari
- Molecular Modeling Lab, Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, Punjab, 147002, India.
| |
Collapse
|
13
|
Galassi A, Ciceri P, Porata G, Iatrino R, Boni Brivio G, Fasulo E, Magagnoli L, Stucchi A, Frittoli M, Cara A, Cozzolino M. Current treatment options for secondary hyperparathyroidism in patients with stage 3 to 4 chronic kidney disease and vitamin D deficiency. Expert Opin Drug Saf 2021; 20:1333-1349. [PMID: 33993809 DOI: 10.1080/14740338.2021.1931117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Secondary hyperparathyroidism (SHPT) represents a complication of chronic kidney disease (CKD). Vitamin D system is altered since early CKD, and vitamin D deficiency is an established trigger of SHPT. Although untreated SHPT may degenerate into tertiary hyperparathyroidism with detrimental consequences in advanced CKD, best treatments for counteracting SHPT from stage 3 CKD are still debated. Enthusiasm on prescription of vitamin D receptor activators (VDRA) in non-dialysis renal patients, has been mitigated by the risk of low bone turnover and positive calcium-phosphate balance. Nutritional vitamin D is now suggested as first-line therapy to treat SHPT with low 25(OH)D insufficiency. However, no high-grade evidence supports the best choice between ergocalciferol, cholecalciferol, and calcifediol (in its immediate or extended-release formulation).Areas covered: The review discusses available data on safety and efficacy of nutritional vitamin D, VDRA and nutritional therapy in replenishing 25(OH)D deficiency and counteracting SHPT in non-dialysis CKD patients.Expert opinion: Best treatment for low 25(OH)D and SHPT remains unknown, due to incomplete understanding of the best homeostatic, as mutable, adaptation of mineral metabolism to CKD progression. Nutritional vitamin D and nutritional therapy appear safest interventions, whenever contextualized with single-patient characteristics. VDRA should be restricted to uncontrolled SHPT by first-line therapy.
Collapse
Affiliation(s)
- Andrea Galassi
- Renal and Dialysis Unit, ASST Santi Paolo E Carlo, Milan, Italy
| | - Paola Ciceri
- Renal Research Laboratory, Department of Nephrology, Dialysis and Renal Transplant, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico and Fondazione D'Amico per La Ricerca Sulle Malattie Renali, Milan, Italy
| | - Giulia Porata
- Renal and Dialysis Unit, ASST Santi Paolo E Carlo, Milan, Italy
| | | | - Giulia Boni Brivio
- Renal and Dialysis Unit, ASST Santi Paolo E Carlo, Milan, Italy.,Department of Health and Science, University of Milan, Milan, Italy
| | - Eliana Fasulo
- Renal and Dialysis Unit, ASST Santi Paolo E Carlo, Milan, Italy
| | - Lorenza Magagnoli
- Renal and Dialysis Unit, ASST Santi Paolo E Carlo, Milan, Italy.,Department of Health and Science, University of Milan, Milan, Italy
| | - Andrea Stucchi
- Renal and Dialysis Unit, ASST Santi Paolo E Carlo, Milan, Italy
| | - Michela Frittoli
- Renal and Dialysis Unit, ASST Santi Paolo E Carlo, Milan, Italy.,Department of Health and Science, University of Milan, Milan, Italy
| | - Anila Cara
- Renal and Dialysis Unit, ASST Santi Paolo E Carlo, Milan, Italy.,Department of Health and Science, University of Milan, Milan, Italy
| | - Mario Cozzolino
- Renal and Dialysis Unit, ASST Santi Paolo E Carlo, Milan, Italy.,Department of Health and Science, University of Milan, Milan, Italy
| |
Collapse
|
14
|
Huang A, Binmahfouz L, Hancock DP, Anderson PH, Ward DT, Conigrave AD. Calcium-Sensing Receptors Control CYP27B1-Luciferase Expression: Transcriptional and Posttranscriptional Mechanisms. J Endocr Soc 2021; 5:bvab057. [PMID: 34337274 PMCID: PMC8317635 DOI: 10.1210/jendso/bvab057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Indexed: 12/19/2022] Open
Abstract
25-hydroxyvitamin D 1α-hydroxylase (encoded by CYP27B1), which catalyzes the synthesis of 1,25-dihydroxyvitamin D3, is subject to negative or positive modulation by extracellular Ca2+ (Ca2+o) depending on the tissue. However, the Ca2+ sensors and underlying mechanisms are unidentified. We tested whether calcium-sensing receptors (CaSRs) mediate Ca2+o-dependent control of 1α-hydroxylase using HEK-293 cells stably expressing the CaSR (HEK-CaSR cells). In HEK-CaSR cells, but not control HEK-293 cells, cotransfected with reporter genes for CYP27B1-Photinus pyralis (firefly) luciferase and control Renilla luciferase, an increase in Ca2+o from 0.5mM to 3.0mM induced a 2- to 3-fold increase in firefly luciferase activity as well as mRNA and protein levels. Surprisingly, firefly luciferase was specifically suppressed at Ca2+o ≥ 5.0mM, demonstrating biphasic Ca2+o control. Both phases were mediated by CaSRs as revealed by positive and negative modulators. However, Ca2+o induced simple monotonic increases in firefly luciferase and endogenous CYP27B1 mRNA levels, indicating that the inhibitory effect of high Ca2+o was posttranscriptional. Studies with inhibitors and the CaSR C-terminal mutant T888A identified roles for protein kinase C (PKC), phosphorylation of T888, and extracellular regulated protein kinase (ERK)1/2 in high Ca2+o-dependent suppression of firefly luciferase. Blockade of both PKC and ERK1/2 abolished Ca2+o-stimulated firefly luciferase, demonstrating that either PKC or ERK1/2 is sufficient to stimulate the CYP27B1 promoter. A key CCAAT box (−74 bp to −68 bp), which is regulated downstream of PKC and ERK1/2, was required for both basal transcription and Ca2+o-mediated transcriptional upregulation. The CaSR mediates Ca2+o-dependent transcriptional upregulation of 1α-hydroxylase and an additional CaSR-mediated mechanism is identified by which Ca2+o can promote luciferase and possibly 1α-hydroxylase breakdown.
Collapse
Affiliation(s)
- Alice Huang
- School of Life and Environmental Sciences, Charles Perkins Centre (D17), University of Sydney, NSW 2006Australia
| | - Lenah Binmahfouz
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Dale P Hancock
- School of Life and Environmental Sciences, Charles Perkins Centre (D17), University of Sydney, NSW 2006Australia
| | - Paul H Anderson
- Clinical and Health Sciences, Health and Biomedical Innovation, University of South Australia, Adelaide, SA, 5001, Australia
| | - Donald T Ward
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Arthur D Conigrave
- School of Life and Environmental Sciences, Charles Perkins Centre (D17), University of Sydney, NSW 2006Australia
| |
Collapse
|
15
|
Vitamin D and its analogs as anticancer and anti-inflammatory agents. Eur J Med Chem 2020; 207:112738. [DOI: 10.1016/j.ejmech.2020.112738] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/29/2020] [Accepted: 08/06/2020] [Indexed: 12/17/2022]
|
16
|
Annamalai C, Ganesh RN, Viswanathan P. Ferrotoxicity and its amelioration by endogenous vitamin D in experimental acute kidney injury. Exp Biol Med (Maywood) 2020; 245:1474-1489. [PMID: 32741217 PMCID: PMC7553091 DOI: 10.1177/1535370220946271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
This work provides in-depth insights on catalytic iron-induced cytotoxicity and the resultant triggering of endogenous vitamin D synthesis in experimental acute kidney injury. Our results reveal significantly elevated levels of catalytic iron culminating in oxidant-mediated renal injury and a concomitant increase in 1,25-dihdyroxyvitamin D3 levels. Also, changes in other iron-related proteins including transferrin, ferritin, and hepcidin were observed both in the serum as well as in their mRNA expression. We consider all these findings vital since no connection between catalytic iron and vitamin D has been established so far. Furthermore, we believe that this work provides new and interesting results, with catalytic iron emerging as an important target in ameliorating renal cellular injury, possibly by timely administration of vitamin D. It also needs to be seen if these observations made in rats could be translated to humans by means of robust clinical trials.
Collapse
Affiliation(s)
- Chandrashekar Annamalai
- Renal Research Lab, Centre for Biomedical Research, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632 014, India
| | - Rajesh N Ganesh
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry 605 006, India
| | - Pragasam Viswanathan
- Renal Research Lab, Centre for Biomedical Research, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632 014, India
- Pragasam Viswanathan.
| |
Collapse
|
17
|
De Paolis E, Scaglione GL, De Bonis M, Minucci A, Capoluongo E. CYP24A1 and SLC34A1 genetic defects associated with idiopathic infantile hypercalcemia: from genotype to phenotype. Clin Chem Lab Med 2020; 57:1650-1667. [PMID: 31188746 DOI: 10.1515/cclm-2018-1208] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/22/2019] [Indexed: 02/07/2023]
Abstract
Loss of function mutations in the CYP24A1 gene, involved in vitamin D catabolism and in calcium homeostasis, are known to be the genetic drivers of both idiopathic infantile hypercalcemia (IIH) and adult renal stone disease. Recently, also defects in the SLC34A1 gene, encoding for the renal sodium-phosphate transporter NaPi-IIa, were associated with the disease. IIH typically affects infants and pediatric patients with a syndrome characterized by severe hypercalcemia, hypercalciuria, suppressed parathyroid hormone level and nephrolithiasis. In SLC34A1 mutated carriers, hypophosphatemia is also a typical biochemical tract. IIH may also persist undiagnosed into adulthood, causing an increased risk of nephrocalcinosis and renal complication. To note, a clinical heterogeneity characterizes IIH manifestation, principally due to the controversial gene-dose effect and, to the strong influence of environmental factors. The present review is aimed to provide an overview of the current molecular findings on the IIH disorder, giving a comprehensive description of the association between genotype and biochemical and clinical phenotype of the affected patients. We also underline that patients may benefit from genetic testing into a targeted diagnostic and therapeutic workflow.
Collapse
Affiliation(s)
- Elisa De Paolis
- Laboratory of Molecular Diagnostics and Genomics, Teaching and Research Hospital "Fondazione Policlinico Agostino Gemelli" - IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Giovanni Luca Scaglione
- Laboratory of Molecular Oncology, "Fondazione Giovanni Paolo II", Catholic University of Sacred Heart, Campobasso, Italy
| | - Maria De Bonis
- Laboratory of Molecular Diagnostics and Genomics, Teaching and Research Hospital "Fondazione Policlinico Agostino Gemelli" - IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Angelo Minucci
- Laboratory of Molecular Diagnostics and Genomics, Teaching and Research Hospital "Fondazione Policlinico Agostino Gemelli" - IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Ettore Capoluongo
- Laboratory of Molecular Diagnostics and Genomics, Teaching and Research Hospital "Fondazione Policlinico Agostino Gemelli" - IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
18
|
Annamalai C, Ganesh RN, Viswanathan P. Ferrotoxicity and its amelioration by endogenous vitamin D in experimental acute kidney injury. Exp Biol Med (Maywood) 2020. [DOI: https://doi.org/10.1177/1535370220946271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Acute kidney injury causes significant morbidity and mortality. This experimental animal study investigated the simultaneous impact of iron and vitamin D on acute kidney injury induced by iohexol, an iodinated, non-ionic monomeric radiocontrast agent in Wistar rats. Out of 36 healthy male Wistar rats, saline was injected into six control rats (group 1) and iohexol into the remaining 30 experimental rats (groups 2 to 6 comprising six rats each). Biochemical, renal histological changes, and gene expression of iron-regulating proteins and 1 α-hydroxylase were analyzed. Urinary neutrophil gelatinase-associated lipocalin (NGAL), serum creatinine, urine protein, serum and urine catalytic iron, 25-hydroxyvitamin D3, 1,25-dihydroxyvitamin D3, and tissue lipid peroxidation were assayed. Rats injected with iohexol showed elevated urinary NGAL (11.94 ± 6.79 ng/mL), serum creatinine (2.92 ± 0.91 mg/dL), and urinary protein levels (11.03 ± 9.68 mg/mg creatinine) together with histological evidence of tubular injury and iron accumulation. Gene expression of iron-regulating proteins and 1 α-hydroxylase was altered. Serum and urine catalytic iron levels were elevated (0.57 ± 0.17; 48.95 ± 29.13 µmol/L) compared to controls (0.49 ± 0.04; 20.7 ± 2.62 µmol/L, P < 0.001). Urine catalytic iron positively correlated with tissue peroxidation (r = 0.469, CI 0.122 to 0.667, P = 0.004) and urinary NGAL (r = 0.788, CI 0.620 to 0.887, P < 0.001). 25-hydroxyvitamin D3 (61.58 ± 9.60 ng/mL) and 1,25-dihydroxyvitamin D3 (50.44 ± 19.76 pg/mL) levels increased simultaneously. In a multivariate linear regression analysis, serum iron, urine catalytic iron, and tissue lipid peroxidation independently and positively predicted urinary NGAL, an acute kidney injury biomarker. This study highlights the nephrotoxic potential of catalytic iron besides demonstrating a concurrent induction of vitamin D endogenously for possible renoprotection in acute kidney injury.Impact statementThis work provides in-depth insights on catalytic iron-induced cytotoxicity and the resultant triggering of endogenous vitamin D synthesis in experimental acute kidney injury. Our results reveal significantly elevated levels of catalytic iron culminating in oxidant-mediated renal injury and a concomitant increase in 1,25-dihdyroxyvitamin D3 levels. Also, changes in other iron-related proteins including transferrin, ferritin, and hepcidin were observed both in the serum as well as in their mRNA expression. We consider all these findings vital since no connection between catalytic iron and vitamin D has been established so far. Furthermore, we believe that this work provides new and interesting results, with catalytic iron emerging as an important target in ameliorating renal cellular injury, possibly by timely administration of vitamin D. It also needs to be seen if these observations made in rats could be translated to humans by means of robust clinical trials.
Collapse
Affiliation(s)
- Chandrashekar Annamalai
- Renal Research Lab, Centre for Biomedical Research, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632 014, India
| | - Rajesh N Ganesh
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry 605 006, India
| | - Pragasam Viswanathan
- Renal Research Lab, Centre for Biomedical Research, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632 014, India
| |
Collapse
|
19
|
Kikuyama T, Susa T, Tamamori-Adachi M, Iizuka M, Akimoto M, Okinaga H, Fujigaki Y, Uchida S, Shibata S, Okazaki T. 25(OH)D 3 stimulates the expression of vitamin D target genes in renal tubular cells when Cyp27b1 is abrogated. J Steroid Biochem Mol Biol 2020; 199:105593. [PMID: 31945466 DOI: 10.1016/j.jsbmb.2020.105593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/28/2022]
Abstract
Recently, it was reported that 25(OH)D3 (25D3) has physiological bioactivity in certain tissues derived from Cyp27b1 knockout mice. To investigate the function of 25D3 in the kidney as an informational crossroad of various calciotropic substances, we employed the CRISPR-Cas9 system to knock out Cyp27b1 in the mouse renal distal tubular mDCT cell line. Unlike the previously reported mice in which Cyp27b1 was targeted systemically, Cyp27b1 knockout mDCT cells did not produce any measurable 1α,25(OH)2D3 (1,25D3) after 25D3 administration. As was seen with treatment of Cyp27b1 knockout mDCT cells with ≥10-8 M of 1,25D3, the administration of 10-7 M of 25D3 translocated the vitamin D3 receptor (VDR) into the nucleus and promoted the expression of the representative 1,25D3-responsive gene Cyp24a1. The exhaustive target gene profiles of 25D3 were similar to those of 1,25D3. Subsequently, we confirmed that 25D3 induced the expression of the calcium reabsorption-related gene calbindin-D9K, in a way similar to 1,25D3. We also found that 1,25D3 and 25D3 induced the expression of the megalin gene. A chromatin immunoprecipitation assay identified two vitamin D response elements in the upstream region of the megalin gene that seemed to contribute to its expression. Together, we surmise that the ability of 25D3 to stimulate VDR target genes may provide a novel perspective for its role in certain tissues.
Collapse
Affiliation(s)
- Takahiro Kikuyama
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Takao Susa
- Department of Biochemistry, Teikyo University School of Medicine, Tokyo, Japan.
| | | | - Masayoshi Iizuka
- Department of Biochemistry, Teikyo University School of Medicine, Tokyo, Japan
| | - Miho Akimoto
- Department of Biochemistry, Teikyo University School of Medicine, Tokyo, Japan
| | - Hiroko Okinaga
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Yoshihide Fujigaki
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Shunya Uchida
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan.
| | - Tomoki Okazaki
- Department of Biochemistry, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
20
|
Chukir T, Liu Y, Hoffman K, Bilezikian JP, Farooki A. Calcitriol Elevation Is Associated with a Higher Risk of Refractory Hypercalcemia of Malignancy in Solid Tumors. J Clin Endocrinol Metab 2020; 105:5679485. [PMID: 31841590 PMCID: PMC7067545 DOI: 10.1210/clinem/dgz278] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/13/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Hypercalcemia of malignancy (HCM) is a common complication of advanced cancer. PTH-independent HCM may be mediated through different mechanisms: (1) humoral HCM, caused by the secretion of PTH-related peptide (PTHrP), (2) local osteolysis resulting from metastatic lesions, and (3) calcitriol-mediated hypercalcemia. Calcitriol-mediated HCM in patients with nonlymphomatous solid tumors is thought to be rare. METHODS We performed a retrospective chart review from 2008 to 2017 to characterize further patients at our institution with solid tumors who had HCM with concomitant elevations in calcitriol. Patients with PTH-dependent hypercalcemia and patients with evidence of granulomatous disease were excluded, as were patients with hematologic malignancies. We hypothesized that patients with HCM and elevated calcitriol levels would respond less favorably to treatment with antiresorptive therapy compared with patients with HCM but without calcitriol elevation. We also aimed to assess mortality and determine if PTHrP and phosphorus levels correlate with calcitriol because both factors may alter calcitriol levels. RESULTS Of 101 eligible patients, calcitriol was elevated in 45 (45%). PTHrP was elevated in 76% of patients with elevated calcitriol compared with 52% of patients without calcitriol elevation. The mean PTHrP value did not differ between patients with HCM and elevated calcitriol (36.3 ± 22 pg/mL) and those without calcitriol elevation (37.4 ± 19 pg/mL). Those with elevated calcitriol levels generally did not respond completely to antiresorptive treatment (80% incomplete response rate), whereas most patients without an elevation in calcitriol responded well to antiresorptive treatment (78% response rate: P < .001). There was no significant difference in the percentage of patients with metastatic bone disease among the 2 groups (49% vs. 55%, respectively). There was no difference in mortality between the 2 groups (P = .14). A weak but significant negative correlation was found between phosphorus and calcitriol (Pearson r = -0.261, P = .016). This correlation was only significant in patients without calcitriol elevation (Pearson r = -0.4, P = .0082). Also, a significant negative correlation was found between PTHrP and phosphorus, again only in patients without calcitriol elevation. DISCUSSION In the setting of HCM, patients with calcitriol elevation are much less likely to respond to antiresorptive therapy than patients without calcitriol elevation. Because calcitriol elevation did not appear to be correlated with hypophosphatemia or elevated PTHrP, it would appear that calcitriol production under these conditions is autonomous, and not subject to normal physiological controls. These observations indicate that calcitriol elevations in patients with HCM have clinical significance.
Collapse
Affiliation(s)
- Tariq Chukir
- Endocrinology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, NewYork-Presbyterian/Weill Cornell Medical Center, New York, New York
- Correspondence and Reprint Requests: Tariq Chukir, MD, NewYork-Presbyterian Hospital/Weill Cornell Medical Center, New York, New York 10065. E-mail:
| | - Yi Liu
- Department of Medicine, Lahey Hospital and Medical Center, Burlington, Massachusetts
| | - Katherine Hoffman
- Division of Biostatistics and Epidemiology, Department of Healthcare Policy and Research, Weill Cornell Medicine, New York Presbyterian Hospital, New York, New York
| | - John P Bilezikian
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Azeez Farooki
- Endocrinology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
21
|
Ramakrishnan A, Kannan S, Warrier R, Nadig MR, Zaidi SN, Kudari A. Diagnosis of TIO: Is Serum FGF23 Measurement always the Answer? Indian J Endocrinol Metab 2020; 24:230-231. [PMID: 32699802 PMCID: PMC7333754 DOI: 10.4103/ijem.ijem_640_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 11/04/2022] Open
Affiliation(s)
- Anantharaman Ramakrishnan
- MAGNA CODE Clinics for Obesity, Diabetes and Endocrinology, Narayana Health City Bangalore, Bengaluru, Karnataka, India
| | - Subramanian Kannan
- Department of Endocrinology, Diabetes and Metabolism, Narayana Health City Bangalore, Bengaluru, Karnataka, India
| | - Rohit Warrier
- MAGNA CODE Clinics for Obesity, Diabetes and Endocrinology, Narayana Health City Bangalore, Bengaluru, Karnataka, India
| | - Murali R Nadig
- Chief Consultant Nuclear Medicine, Physician and Director, NewMedd Diagnostics, Bengaluru, Karnataka, India
| | - Shaesta Naseem Zaidi
- Department of Pathology and Laboratory Services, Narayana Health City, Bengaluru, Karnataka, India
| | - Ashwinikumar Kudari
- Surgical Gastroenterology and Robotic Surgery, Narayana Health City, Bengaluru, Karnataka, India
| |
Collapse
|
22
|
Pal R, Bhadada SK, Singhare A, Bhansali A, Kamalanathan S, Chadha M, Chauhan P, Sood A, Dhiman V, Sharma DC, Saikia UN, Chatterjee D, Agashe V. Tumor-induced osteomalacia: experience from three tertiary care centers in India. Endocr Connect 2019; 8:266-276. [PMID: 30726771 PMCID: PMC6410764 DOI: 10.1530/ec-18-0552] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 12/17/2022]
Abstract
Tumor-induced osteomalacia (TIO) is a rare paraneoplastic syndrome characterized by recalcitrant hypophosphatemia. Reports from the Indian subcontinent are scarce, with most being single center experiences involving few patients. Herein, we conducted a retrospective analysis of 30 patients of TIO diagnosed at three tertiary care hospitals in India. Patients with persistent hypophosphatemia (despite correction of hypovitaminosis D), normocalcemia, elevated alkaline phosphatase, low TmP/GFR and elevated or 'inappropriately normal' FGF23 levels were labeled as having TIO. They were sequentially subjected to functional followed by anatomical imaging. Patients with a well-localized tumor underwent excision; others were put on phosphorous and calcitriol supplementation. The mean age at presentation was 39.6 years with female:male ratio of 3:2. Bone pain (83.3%) and proximal myopathy (70%) were the chief complaints; 40% of cases had fractures. The mean delay in diagnosis was 3.8 years. Tumors were clinically detectable in four patients (13.3%). The mean serum phosphate was 0.50 mmol/L with a median serum FGF23 level of 518 RU/mL. Somatostatin receptor-based scintigraphy was found to be superior to FDG-PET in tumor localization. Lower extremities were the most common site of the tumor (72%). Tumor size was positively correlated with serum FGF23 levels. Twenty-two patients underwent tumor resection and 16 of them had phosphaturic mesenchymal tumors. Surgical excision led to cure in 72.7% of patients whereas disease persistence and disease recurrence were seen in 18.2% and 9.1% of cases, respectively. At the last follow-up, serum phosphate in the surgically treated group was significantly higher than in the medically managed group.
Collapse
Affiliation(s)
- Rimesh Pal
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Sanjay Kumar Bhadada
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
- Correspondence should be addressed to S K Bhadada:
| | - Awesh Singhare
- Department of Endocrinology, P D Hinduja Hospital and Medical Research Centre, Mumbai, India
| | - Anil Bhansali
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Sadishkumar Kamalanathan
- Department of Endocrinology, Jawaharlal Institute of Post Graduate Medical Education and Research, Pondicherry, India
| | - Manoj Chadha
- Department of Endocrinology, P D Hinduja Hospital and Medical Research Centre, Mumbai, India
| | - Phulrenu Chauhan
- Department of Endocrinology, P D Hinduja Hospital and Medical Research Centre, Mumbai, India
| | - Ashwani Sood
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Vandana Dhiman
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Uma Nahar Saikia
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Debajyoti Chatterjee
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Vikas Agashe
- Department of Orthopaedics, P D Hinduja Hospital and Medical Research Centre, Mumbai, India
| |
Collapse
|
23
|
Lu CL, Yeih DF, Hou YC, Jow GM, Li ZY, Liu WC, Zheng CM, Lin YF, Shyu JF, Chen R, Huang CY, Lu KC. The Emerging Role of Nutritional Vitamin D in Secondary Hyperparathyroidism in CKD. Nutrients 2018; 10:nu10121890. [PMID: 30513912 PMCID: PMC6316278 DOI: 10.3390/nu10121890] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/22/2018] [Accepted: 11/23/2018] [Indexed: 12/18/2022] Open
Abstract
In chronic kidney disease (CKD), hyperphosphatemia induces fibroblast growth factor-23 (FGF-23) expression that disturbs renal 1,25-dihydroxy vitamin D (1,25D) synthesis; thereby increasing parathyroid hormone (PTH) production. FGF-23 acts on the parathyroid gland (PTG) to increase 1α-hydroxylase activity and results in increase intra-gland 1,25D production that attenuates PTH secretion efficiently if sufficient 25D are available. Interesting, calcimimetics can further increase PTG 1α-hydroxylase activity that emphasizes the demand for nutritional vitamin D (NVD) under high PTH status. In addition, the changes in hydroxylase enzyme activity highlight the greater parathyroid 25-hydroxyvitmain D (25D) requirement in secondary hyperparathyroidism (SHPT); the higher proportion of oxyphil cells as hyperplastic parathyroid progression; lower cytosolic vitamin D binding protein (DBP) content in the oxyphil cell; and calcitriol promote vitamin D degradation are all possible reasons supports nutritional vitamin D (NVD; e.g., Cholecalciferol) supplement is crucial in SHPT. Clinically, NVD can effectively restore serum 25D concentration and prevent the further increase in PTH level. Therefore, NVD might have the benefit of alleviating the development of SHPT in early CKD and further lowering PTH in moderate to severe SHPT in dialysis patients.
Collapse
Affiliation(s)
- Chien-Lin Lu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| | - Dong-Feng Yeih
- Division of Cardiology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| | - Yi-Chou Hou
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Division of Nephrology, Department of Medicine, Cardinal-Tien Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City 23155, Taiwan.
| | - Guey-Mei Jow
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| | - Zong-Yu Li
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| | - Wen-Chih Liu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Division of Nephrology, Department of Internal Medicine, Tungs' Taichung MetroHarbor Hospital, Taichung City 433, Taiwan.
| | - Cai-Mei Zheng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, New Taipei City 235, Taiwan.
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11103, Taiwan.
| | - Yuh-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, New Taipei City 235, Taiwan.
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11103, Taiwan.
| | - Jia-Fwu Shyu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 114, Taiwan.
| | - Remy Chen
- Kidney Dialysis Center, Kamifukuoka General Hospital, Saitama 356, Japan.
| | - Chung-Yu Huang
- Department of Medicine, Show-Chwan Memorial Hospital, Changhua 50008, Taiwan.
| | - Kuo-Cheng Lu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| |
Collapse
|
24
|
Abstract
Inorganic phosphate (Pi) is essential for signal transduction and cell metabolism, and is also an essential structural component of the extracellular matrix of the skeleton. Pi is sensed in bacteria and yeast at the plasma membrane, which activates intracellular signal transduction to control the expression of Pi transporters and other genes that control intracellular Pi levels. In multicellular organisms, Pi homeostasis must be maintained in the organism and at the cellular level, requiring an endocrine and metabolic Pi-sensing mechanism, about which little is currently known. This Review will discuss the metabolic effects of Pi, which are mediated by Pi transporters, inositol pyrophosphates and SYG1-Pho81-XPR1 (SPX)-domain proteins to maintain cellular phosphate homeostasis in the musculoskeletal system. In addition, we will discuss how Pi is sensed by the human body to regulate the production of fibroblast growth factor 23 (FGF23), parathyroid hormone and calcitriol to maintain serum levels of Pi in a narrow range. New findings on the crosstalk between iron and Pi homeostasis in the regulation of FGF23 expression will also be outlined. Mutations in components of these metabolic and endocrine phosphate sensors result in genetic disorders of phosphate homeostasis, cardiomyopathy and familial basal ganglial calcifications, highlighting the importance of this newly emerging area of research.
Collapse
Affiliation(s)
- Sampada Chande
- Section of Endocrinology and Metabolism, Yale University School of Medicine, New Haven, CT, USA
| | - Clemens Bergwitz
- Section of Endocrinology and Metabolism, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
25
|
Time-varying coefficient of determination to quantify the explanatory power of biomarkers on longitudinal GFR among children with chronic kidney disease. Ann Epidemiol 2018; 28:549-556. [PMID: 29887227 DOI: 10.1016/j.annepidem.2018.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 03/29/2018] [Accepted: 05/07/2018] [Indexed: 01/07/2023]
Abstract
PURPOSE Coefficients of determination (R2) for continuous longitudinal data are typically reported as time constant, if they are reported at all. The widely used mixed model with random intercepts and slopes yields the total outcome variance as a time-varying function. We propose a generalized and intuitive approach based on this variance function to estimate the time-varying predictive power (R2) of a variable on outcome levels and changes. METHODS Using longitudinal estimated glomerular filtration rate (eGFR) from the Chronic Kidney Disease in Children Study, linear mixed models characterized the R2 for two chronic kidney disease (CKD) risk factors measured at baseline: a traditional marker (proteinuria) and a novel marker (fibroblast growth factor 23 [FGF23]). RESULTS Time-varying R2 divulged different disease processes by risk factor and diagnoses. Among children with glomerular CKD, time-varying R2 for proteinuria had significant upward trends, suggesting increasing power to predict eGFR change, but crossed with FGF23, which was higher up to 2.5 years from baseline. In contrast, among those with nonglomerular CKD, proteinuria explained more than FGF23 at all times, and time-varying R2 for each risk factor was not substantially different from time-constant estimates. CONCLUSIONS Proteinuria and FGF23 explained substantial eGFR variability over time. Time-varying R2 can characterize predictive roles of risk factors on disease progression, overcome limitations of time-constant estimates, and are easily derived from mixed effects models.
Collapse
|
26
|
Marik B, Bagga A, Sinha A, Hari P, Sharma A. Genetics of Refractory Rickets: Identification of Novel PHEX Mutations in Indian Patients and a Literature Update. J Pediatr Genet 2018; 7:47-59. [PMID: 29707405 PMCID: PMC5916800 DOI: 10.1055/s-0038-1624577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 12/25/2017] [Indexed: 10/18/2022]
Abstract
Refractory rickets is a genetic disorder that cannot be treated by vitamin D supplementation and adequate dietary calcium and phosphorus. Hereditary hypophosphatemic rickets is one of the major forms of refractory rickets in Indian children and caused due to mutations in the PHEX , FGF23 , DMP1 , ENPP1 , and SLC34A3 genes. This is the first study in India on a large number of patients reporting on mutational screening of the PHEX gene. Direct sequencing in 37 patients with refractory rickets revealed eight mutations in 13 patients of which 1 was nonsense, 2 were deletions, 1 was a deletion-insertion, and 4 were missense mutations. Of these mutations, four (c.566_567 delAG, c.651_654delACAT, c.1337delinsAATAA, and c.2048T > A) were novel mutations. This article discusses the mutations in Indian patients, collates information on the genetic causes of refractory rickets, and emphasizes the significance of genetic testing for precise diagnosis, timely treatment, and management of the condition, especially in developing countries.
Collapse
Affiliation(s)
- Binata Marik
- Division of Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Arvind Bagga
- Department of Paediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Aditi Sinha
- Department of Paediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Pankaj Hari
- Department of Paediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Arundhati Sharma
- Division of Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
27
|
The intestinal phosphate transporter NaPi-IIb (Slc34a2) is required to protect bone during dietary phosphate restriction. Sci Rep 2017; 7:11018. [PMID: 28887454 PMCID: PMC5591270 DOI: 10.1038/s41598-017-10390-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 08/07/2017] [Indexed: 02/04/2023] Open
Abstract
NaPi-IIb/Slc34a2 is a Na+-dependent phosphate transporter that accounts for the majority of active phosphate transport into intestinal epithelial cells. Its abundance is regulated by dietary phosphate, being high during dietary phosphate restriction. Intestinal ablation of NaPi-IIb in mice leads to increased fecal excretion of phosphate, which is compensated by enhanced renal reabsorption. Here we compared the adaptation to dietary phosphate of wild type (WT) and NaPi-IIb−/− mice. High phosphate diet (HPD) increased fecal and urinary excretion of phosphate in both groups, though NaPi-IIb−/− mice still showed lower urinary excretion than WT. In both genotypes low dietary phosphate (LDP) resulted in reduced fecal excretion and almost undetectable urinary excretion of phosphate. Consistently, the expression of renal cotransporters after prolonged LDP was similar in both groups. Plasma phosphate declined more rapidly in NaPi-IIb−/− mice upon LDP, though both genotypes had comparable levels of 1,25(OH)2vitamin D3, parathyroid hormone and fibroblast growth factor 23. Instead, NaPi-IIb−/− mice fed LDP had exacerbated hypercalciuria, higher urinary excretion of corticosterone and deoxypyridinoline, lower bone mineral density and higher number of osteoclasts. These data suggest that during dietary phosphate restriction NaPi-IIb-mediated intestinal absorption prevents excessive demineralization of bone as an alternative source of phosphate.
Collapse
|
28
|
Batacchi Z, Robinson-Cohen C, Hoofnagle AN, Isakova T, Kestenbaum B, Martin KJ, Wolf MS, de Boer IH. Effects of Vitamin D 2 Supplementation on Vitamin D 3 Metabolism in Health and CKD. Clin J Am Soc Nephrol 2017; 12:1498-1506. [PMID: 28768705 PMCID: PMC5586563 DOI: 10.2215/cjn.00530117] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 05/25/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND OBJECTIVES Vitamin D supplements are prescribed to correct low circulating concentrations of 25-hydroxyvitamin D. In CKD, vitamin D metabolism is complicated by decreased conversion of 25-hydroxyvitamin D to 1,25-dihydroxyvitamin D by CYP27B1 and possibly decreased conversion of 25-hydroxyvitamin D to 24,25-dihydroxyvitamin D by CYP24A1. The aim of this study was to determine the effects of vitamin D2 supplementation on vitamin D metabolism in health and CKD. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We conducted a treatment-only intervention study of 25 individuals with CKD (eGFR<60 ml/min per 1.73 m2) and 44 individuals without CKD from three academic centers, all with screening 25-hydroxyvitamin D <30 ng/ml. Each participant was prescribed vitamin D2 (ergocalciferol) 50,000 IU orally twice weekly for 5 weeks. We tested whether changes in plasma concentrations of vitamin D metabolites and vitamin D metabolic ratios differed by CKD status. Plasma 1,25-dihydroxyvitamin D3-to-25-hydroxyvitamin D3 ratio and 24,25-dihydroxyvitamin D3-to-25-hydroxyvitamin D3 ratio were calculated as estimates of CYP27B1 and CYP24A1 function, respectively. RESULTS With treatment, plasma 25-hydroxyvitamin D2 and total 25-hydroxyvitamin D concentrations increased similarly for participants with and without CKD. For participants without CKD, 1,25-dihydroxyvitamin D2 increased (2.8±1.3-32.9±1.4 pg/ml), whereas 1,25-dihydroxyvitamin D3 decreased (45.6±1.9-14.6±1.9 pg/ml), resulting in no significant change in total 1,25-dihydroxyvitamin D; 1,25-dihydroxyvitamin D3-to-25-hydroxyvitamin D3 ratio decreased (3.0±0.2-1.7±0.2 pg/ng), and 24,25-dihydroxyvitamin D3-to-25-hydroxyvitamin D3 ratio increased (115.7±7.8-195.2±7.9 pg/ng). Individuals with CKD had lower baseline levels and smaller changes in magnitude for 1,25-dihydroxyvitamin D2 (2.1±1.6-24.4±1.6 pg/ml; P interaction =0.01), 1,25-dihydroxyvitamin D3-to-25-hydroxyvitamin D3 ratio (1.8±0.2-1.1±0.2 pg/ng; P interaction =0.05), and 24,25-dihydroxyvitamin D3-to-25-hydroxyvitamin D3 ratio (72.0±9.1-110.3±9.3 pg/ng; P interaction <0.001). Fibroblast growth factor-23 and parathyroid hormone were not significantly changed in either group. CONCLUSIONS Vitamin D2 supplementation decreases conversion of 25-hydroxyvitamin D3 to 1,25-dihydroxyvitamin D3 and induces vitamin D3 catabolism as evidenced by changes in D3 metabolites and vitamin D metabolic ratios. These effects occur without significant changes in fibroblast growth factor-23 or parathyroid hormone and are blunted in CKD. PODCAST This article contains a podcast at https://www.asn-online.org/media/podcast/CJASN/2017_08_02_CJASNPodcast_17_09.mp3.
Collapse
Affiliation(s)
- Zona Batacchi
- Divisions of Metabolism, Endocrinology and Nutrition and
- Kidney Research Institute, and
| | | | - Andrew N. Hoofnagle
- Divisions of Metabolism, Endocrinology and Nutrition and
- Kidney Research Institute, and
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Tamara Isakova
- Division of Nephrology and Hypertension, Department of Medicine and
- Center for Translational Metabolism and Health, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Kevin J. Martin
- Division of Nephrology, St. Louis University, St. Louis, Missouri
| | - Myles S. Wolf
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina; and
| | - Ian H. de Boer
- Nephrology
- Kidney Research Institute, and
- Veterans Affairs Puget Sound Health Care System, Seattle, Washington
| |
Collapse
|
29
|
Zhou S, Glowacki J. Chronic kidney disease and vitamin D metabolism in human bone marrow-derived MSCs. Ann N Y Acad Sci 2017; 1402:43-55. [PMID: 28926112 PMCID: PMC5659722 DOI: 10.1111/nyas.13464] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/09/2017] [Accepted: 08/09/2017] [Indexed: 12/18/2022]
Abstract
Vitamin D that is synthesized in the skin or is ingested undergoes sequential steps of metabolic activation via a cascade of cytochrome P450 enzymatic hydroxylations in the liver and kidney to produce 1α,25-dihydroxyvitamin D (1α,25(OH)2 D). There are many tissues that are able to synthesize 1α,25(OH)2 D, but the biological significance of extrarenal hydroxylases is unresolved. Human marrow-derived mesenchymal stem cells (marrow stromal cells, hMSCs) give rise to osteoblasts, and their differentiation is stimulated by 1α,25(OH)2 D. In addition to being targets of 1α,25(OH)2 D, hMSCs can synthesize it; on the basis of those observations, we further examined the local autocrine/paracrine role of vitamin D metabolism in osteoblast differentiation. Research with hMSCs from well-characterized subjects provides an innovative opportunity to evaluate the effects of clinical attributes on the regulation of hMSCs. Like the renal 1α-hydroxylase, the enzyme in hMSCs is constitutively decreased with age and chronic kidney disease (CKD); both are regulated by PTH1-34, insulin-like growth factor 1, calcium, 1α,25(OH)2 D, 25(OH)D, and fibroblast growth factor 23. CKD is associated with impaired renal biosynthesis of 1α,25(OH)2 D, low bone mass, and increased fracture risk. Studies with hMSCs from CKD patients or aged subjects indicate that circulating 25(OH)D may have an important role in osteoblast differentiation on vitamin D metabolism and action in hMSCs.
Collapse
Affiliation(s)
- Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Julie Glowacki
- Department of Orthopedic Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
30
|
Deviations from the expected relationship between serum FGF23 and other markers in children with CKD: a cross-sectional study. BMC Nephrol 2017; 18:204. [PMID: 28659167 PMCID: PMC5490085 DOI: 10.1186/s12882-017-0623-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 06/19/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND High levels of fibroblast growth factor-23 (FGF23) are associated with mortality. In chronic kidney disease (CKD), FGF23 levels rise as renal function declines. We analyzed the contribution of laboratory values to the variance of FGF23 levels in relationship to a curve of expected FGF23 levels for a given GFR. METHODS Following approval by the research ethics boards, we measured FGF23, CysC eGFR, creatinine, urea, albumin, calcium, phosphate, vitamin D metabolites, PTH, alkaline phosphatase, CRP, and venous gases in 141 pediatric CKD patients (45, 37, 32, 13 and 14 CKD stages I, II, III, IV, and V, respectively). Data were expressed as median (25th, 75th percentile). RESULTS FGF23 correlated significantly with CysC, CysC eGFR, PTH, 1.25 (OH)2 vitamin D, phosphate, and pH. The correlation of the latter three remained significant in the multivariate analysis. We calculated a formula for the expected FGF23 value for a given level of eGFR which reads Y = 1295 * e-0.07247*X + 38.35. Deviation by more than 20% from the curve also depended on phosphate, 1.25 (OH)2 vitamin D and pH. CONCLUSIONS Our data emphasize the importance of phosphate and 1.25 (OH)2 vitamin D levels. The impact of acidosis on FGF23 warrants further studies.
Collapse
|
31
|
Dong XL, Cao SS, Zhou LP, Denney L, Wong MS, Feng HT. Ethanol Extract of Fructus ligustri lucidi Increased Circulating 1,25(OH)2D3Levels, but Did Not Improve Calcium Balance in Mature Ovariectomized Rats. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2016; 44:1237-1253. [DOI: 10.1142/s0192415x16500695] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Our previous studies found that different extracts or fractions of Fructus ligustri lucidi (FLL) played different roles in altering the regulation of bone and mineral metabolism in different animal models. The present study was designed to compare the actions of FLL ethanol (EE) and water extracts (WE) on bone and mineral metabolism in a 6-month-old mature ovariectomized (OVX) rat model. Our results showed that FLL extracts did not significantly improve systematic Ca balance in mature OVX rats. However, EE, but not WE treatment, significantly increased serum 1,25(OH)2D3levels in mature OVX rats. An in vitro study using human proximal tubule (HKC-8) cells showed that EE, but not WE, significantly enhanced renal 25-dihydroxyvitamin D3-1[Formula: see text]-hydroxylase (1-OHase) mRNA expressions and simultaneously repressed renal 25-dihydroxyvitamin D3-24-hydroxylase (24-OHase) mRNA expressions. Further investigation indicated that EE could significantly induce the protein expression of 1-OHase, but did not alter 24-OHase expression in HKC-8 cells. Our results demonstrated that EE increased circulating 1,25(OH)2D3levels in OVX rats, possibly via upregulation of renal 1-OHase expressions in renal proximal tubule cells. Our study indicates that FLL is a natural oral agent that could directly regulate renal vitamin D metabolism in vivo and in vitro.
Collapse
Affiliation(s)
- Xiao-Li Dong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, P.R. China
| | - Si-Si Cao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, P.R. China
| | - Li-Ping Zhou
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, P.R. China
| | - Liya Denney
- Nestlé Research Centre, P. O. Box 44, 1000 Lausanne 26, Switzerland
| | - Man-Sau Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, P.R. China
- Shenzhen Research Institute of The Hong Kong Polytechnic University, State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), Shenzhen 518057, P.R. China
| | - Hao-Tian Feng
- Nestlé Research Centre Beijing, No. 5 Dijin Road, Haidian District, Beijing, P.R. China
| |
Collapse
|
32
|
van der Meijden K, van Essen HW, Bloemers FW, Schulten EAJM, Lips P, Bravenboer N. Regulation of CYP27B1 mRNA Expression in Primary Human Osteoblasts. Calcif Tissue Int 2016; 99:164-73. [PMID: 27016371 PMCID: PMC4932130 DOI: 10.1007/s00223-016-0131-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/11/2016] [Indexed: 12/12/2022]
Abstract
The enzyme 1α-hydroxylase (gene CYP27B1) catalyzes the synthesis of 1,25(OH)2D in both renal and bone cells. While renal 1α-hydroxylase is tightly regulated by hormones and 1,25(OH)2D itself, the regulation of 1α-hydroxylase in bone cells is poorly understood. The aim of this study was to investigate in a primary human osteoblast culture whether parathyroid hormone (PTH), fibroblast growth factor 23 (FGF23), calcitonin, calcium, phosphate, or MEPE affect mRNA levels of CYP27B1. Our results show that primary human osteoblasts in the presence of high calcium concentrations increase their CYP27B1 mRNA levels by 1.3-fold. CYP27B1 mRNA levels were not affected by PTH1-34, rhFGF23, calcitonin, phosphate, and rhMEPE. Our results suggest that the regulation of bone 1α-hydroxylase is different from renal 1α-hydroxylase. High calcium concentrations in bone may result in an increased local synthesis of 1,25(OH)2D leading to an enhanced matrix mineralization. In this way, the local synthesis of 1,25(OH)2D may contribute to the stimulatory effect of calcium on matrix mineralization.
Collapse
Affiliation(s)
- K van der Meijden
- Department of Internal Medicine/Endocrinology, VU University Medical Center, Research Institute MOVE, Amsterdam, The Netherlands
| | - H W van Essen
- Department of Clinical Chemistry, VU University Medical Center, Research Institute MOVE, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
| | - F W Bloemers
- Department of Trauma Surgery, VU University Medical Center, Amsterdam, The Netherlands
| | - E A J M Schulten
- Department of Oral and Maxillofacial Surgery/Oral Pathology, VU University Medical Center, Academic Centre for Dentistry Amsterdam (ACTA), Amsterdam, The Netherlands
| | - P Lips
- Department of Internal Medicine/Endocrinology, VU University Medical Center, Research Institute MOVE, Amsterdam, The Netherlands
| | - N Bravenboer
- Department of Clinical Chemistry, VU University Medical Center, Research Institute MOVE, PO Box 7057, 1007 MB, Amsterdam, The Netherlands.
| |
Collapse
|
33
|
Fleet JC, Replogle RA, Reyes-Fernandez P, Wang L, Zhang M, Clinkenbeard EL, White KE. Gene-by-Diet Interactions Affect Serum 1,25-Dihydroxyvitamin D Levels in Male BXD Recombinant Inbred Mice. Endocrinology 2016; 157:470-81. [PMID: 26587785 PMCID: PMC4733130 DOI: 10.1210/en.2015-1786] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
1,25-Dihydroxyvitamin D (1,25[OH]2D) regulates calcium (Ca), phosphate, and bone metabolism. Serum 1,25(OH)2D levels are reduced by low vitamin D status and high fibroblast growth factor 23 (FGF23) levels and increased by low Ca intake and high PTH levels. Natural genetic variation controls serum 25-hydroxyvitamin D (25[OH]D) levels, but it is unclear how it controls serum 1,25(OH)2D or the response of serum 1,25(OH)2D levels to dietary Ca restriction (RCR). Male mice from 11 inbred lines and from 51 BXD recombinant inbred lines were fed diets with either 0.5% (basal) or 0.25% Ca from 4 to 12 weeks of age (n = 8 per line per diet). Significant variation among the lines was found in basal serum 1,25(OH)2D and in the RCR as well as basal serum 25(OH)D and FGF23 levels. 1,25(OH)2D was not correlated to 25(OH)D but was negatively correlated to FGF23 (r = -0.5). Narrow sense heritability of 1,25(OH)2D was 0.67 on the 0.5% Ca diet, 0.66 on the 0.25% Ca diet, and 0.59 for the RCR, indicating a strong genetic control of serum 1,25(OH)2D. Genetic mapping revealed many loci controlling 1,25(OH)2D (seven loci) and the RCR (three loci) as well as 25(OH)D (four loci) and FGF23 (two loci); a locus on chromosome 18 controlled both 1,25(OH)2D and FGF23. Candidate genes underlying loci include the following: Ets1 (1,25[OH]2D), Elac1 (FGF23 and 1,25[OH]2D), Tbc1d15 (RCR), Plekha8 and Lyplal1 (25[OH]D), and Trim35 (FGF23). This report is the first to reveal that serum 1,25(OH)2D levels are controlled by multiple genetic factors and that some of these genetic loci interact with the dietary environment.
Collapse
Affiliation(s)
- James C Fleet
- Departments of Nutrition Science (J.C.F., R.A.R., P.R.-F.) and Statistics (L.W., M.Z.), Purdue University, West Lafayette, Indiana 47907-2059; and Department of Medical and Molecular Genetics (E.L.C., K.E.W.), Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Rebecca A Replogle
- Departments of Nutrition Science (J.C.F., R.A.R., P.R.-F.) and Statistics (L.W., M.Z.), Purdue University, West Lafayette, Indiana 47907-2059; and Department of Medical and Molecular Genetics (E.L.C., K.E.W.), Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Perla Reyes-Fernandez
- Departments of Nutrition Science (J.C.F., R.A.R., P.R.-F.) and Statistics (L.W., M.Z.), Purdue University, West Lafayette, Indiana 47907-2059; and Department of Medical and Molecular Genetics (E.L.C., K.E.W.), Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Libo Wang
- Departments of Nutrition Science (J.C.F., R.A.R., P.R.-F.) and Statistics (L.W., M.Z.), Purdue University, West Lafayette, Indiana 47907-2059; and Department of Medical and Molecular Genetics (E.L.C., K.E.W.), Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Min Zhang
- Departments of Nutrition Science (J.C.F., R.A.R., P.R.-F.) and Statistics (L.W., M.Z.), Purdue University, West Lafayette, Indiana 47907-2059; and Department of Medical and Molecular Genetics (E.L.C., K.E.W.), Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Erica L Clinkenbeard
- Departments of Nutrition Science (J.C.F., R.A.R., P.R.-F.) and Statistics (L.W., M.Z.), Purdue University, West Lafayette, Indiana 47907-2059; and Department of Medical and Molecular Genetics (E.L.C., K.E.W.), Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Kenneth E White
- Departments of Nutrition Science (J.C.F., R.A.R., P.R.-F.) and Statistics (L.W., M.Z.), Purdue University, West Lafayette, Indiana 47907-2059; and Department of Medical and Molecular Genetics (E.L.C., K.E.W.), Indiana University School of Medicine, Indianapolis, Indiana 46202
| |
Collapse
|
34
|
Veldurthy V, Wei R, Campbell M, Lupicki K, Dhawan P, Christakos S. 25-Hydroxyvitamin D₃ 24-Hydroxylase: A Key Regulator of 1,25(OH)₂D₃ Catabolism and Calcium Homeostasis. VITAMINS AND HORMONES 2016; 100:137-50. [PMID: 26827951 DOI: 10.1016/bs.vh.2015.10.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
One of the most pronounced effects of the hormonally active form of vitamin D, 1,25-dihydroxyvitamin D3 (1,25(OH)2D3), is increased synthesis of 25-hydroxyvitamin D3 24-hydroxylase (CYP24A1), the enzyme responsible for the catabolism of 1,25(OH)2D3. Thus, 1,25(OH)2D3 regulates its own metabolism, protecting against hypercalcemia and limiting the levels of 1,25(OH)2D3 in cells. This chapter summarizes the catalytic properties of CYP24A1, the recent data related to the crystal structure of CYP24A1, the findings obtained from the generation of mice deficient for the Cyp24a1 gene as well as recent data identifying a causal role of a genetic defect in CYP24A1 in certain patients with idiopathic infantile hypercalcemia. This chapter also reviews the regulation of renal and placental CYP24A1 as well as the genomic mechanisms, including coactivators, repressors, and epigenetic modification, involved in modulating 1,25(OH)2D3 regulation of CYP24A1. We conclude with future research directions related to this key regulator of 1,25(OH)2D3 catabolism and calcium homeostasis.
Collapse
Affiliation(s)
- Vaishali Veldurthy
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Ran Wei
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Megan Campbell
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Kamil Lupicki
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Puneet Dhawan
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Sylvia Christakos
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA.
| |
Collapse
|
35
|
Interactions of vitamin D and the proximal tubule. Pediatr Nephrol 2016; 31:7-14. [PMID: 25618772 DOI: 10.1007/s00467-015-3050-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/06/2015] [Accepted: 01/07/2015] [Indexed: 01/12/2023]
Abstract
Severe vitamin D deficiency (reduction in serum 25(OH)D concentration) in infants and children can cause features of the Fanconi syndrome, including phosphaturia, glycosuria, aminoaciduria, and renal tubular acidosis. This indicates that vitamin D and its metabolites influence proximal tubule function. Filtered 25(OH)D bound to vitamin D binding protein (DBP) is endocytosed by megalin-cubilin in the apical membrane. Intracellular 25(OH)D is metabolized to 1,25(OH)2D or calcitroic acid by 1-α-hydroxylase or 24-hydroxylase in tubule cell mitochondria. Bone-produced fibroblast growth factor 23 (FGF23) bound to Klotho in tubule cells and intracellular phosphate concentrations are regulators of 1-α-hydroxylase activity and cause proximal tubule phosphaturia. Aminoaciduria occurs when amino acid transporter synthesis is deficient, and 1,25(OH)2D along with retinoic acid up-regulate transporter synthesis by a vitamin D response element in the promoter region of the transporter gene. This review discusses evidence gained from studies in animals or cell lines, as well as from human disorders, that provide insight into vitamin D-proximal tubule interactions.
Collapse
|
36
|
Hernando N, Myakala K, Simona F, Knöpfel T, Thomas L, Murer H, Wagner CA, Biber J. Intestinal Depletion of NaPi-IIb/Slc34a2 in Mice: Renal and Hormonal Adaptation. J Bone Miner Res 2015; 30:1925-37. [PMID: 25827490 DOI: 10.1002/jbmr.2523] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 03/26/2015] [Accepted: 03/27/2015] [Indexed: 11/10/2022]
Abstract
The Na(+) -dependent phosphate-cotransporter NaPi-IIb (SLC34A2) is widely expressed, with intestine, lung, and testis among the organs with highest levels of mRNA abundance. In mice, the intestinal expression of NaPi-IIb is restricted to the ileum, where the cotransporter localizes specifically at the brush border membrane (BBM) and mediates the active transport of inorganic phosphate (Pi). Constitutive full ablation of NaPi-IIb is embryonically lethal whereas the global but inducible removal of the transporter in young mice leads to intestinal loss of Pi and lung calcifications. Here we report the generation of a constitutive but intestinal-specific NaPi-IIb/Slc34a2-deficient mouse model. Constitutive intestinal ablation of NaPi-IIb results in viable pups with normal growth. Homozygous mice are characterized by fecal wasting of Pi and complete absence of Na/Pi cotransport activity in BBM vesicles (BBMVs) isolated from ileum. In contrast, the urinary excretion of Pi is reduced in these animals. The plasma levels of Pi are similar in wild-type and NaPi-IIb-deficient mice. In females, the reduced phosphaturia associates with higher expression of NaPi-IIa and higher Na/Pi cotransport activity in renal BBMVs, as well as with reduced plasma levels of intact FGF-23. A similar trend is found in males. Thus, NaPi-IIb is the only luminal Na(+) -dependent Pi transporter in the murine ileum and its absence is fully compensated for in adult females by a mechanism involving the bone-kidney axis. The contribution of this mechanism to the adaptive response is less apparent in adult males.
Collapse
Affiliation(s)
- Nati Hernando
- Institute of Physiology and Zurich Center for Integrative Human Physiology (ZIHP). University of Zurich, Zurich, Switzerland
| | - Komuraiah Myakala
- Institute of Physiology and Zurich Center for Integrative Human Physiology (ZIHP). University of Zurich, Zurich, Switzerland
| | - Fabia Simona
- Institute of Physiology and Zurich Center for Integrative Human Physiology (ZIHP). University of Zurich, Zurich, Switzerland
| | - Thomas Knöpfel
- Institute of Physiology and Zurich Center for Integrative Human Physiology (ZIHP). University of Zurich, Zurich, Switzerland
| | - Linto Thomas
- Institute of Physiology and Zurich Center for Integrative Human Physiology (ZIHP). University of Zurich, Zurich, Switzerland
| | - Heini Murer
- Institute of Physiology and Zurich Center for Integrative Human Physiology (ZIHP). University of Zurich, Zurich, Switzerland
| | - Carsten A Wagner
- Institute of Physiology and Zurich Center for Integrative Human Physiology (ZIHP). University of Zurich, Zurich, Switzerland
| | - Jürg Biber
- Institute of Physiology and Zurich Center for Integrative Human Physiology (ZIHP). University of Zurich, Zurich, Switzerland
| |
Collapse
|
37
|
Abstract
Substances historically thought to cause direct vascular injury in laboratory animals are a heterogeneous group of toxic agents with varied mechanisms of action. Morphologically, the reviewed agents can be broadly categorized into those targeting endothelial cell (ECs) and those targeting smooth muscle cells (SMCs). Anticancer drugs, immunosuppressants, and heavy metals are targeting primarily ECs while allylamine, β-aminopropionitrile, and mitogen-activated protein kinase kinase inhibitors affect mainly SMCs. It is now recognized that the pathogenicity of some of these agents is often mediated through intermediary events, particularly vasoconstriction. There are clear similarities in the clinical and microscopic findings associated with many of these agents in animals and man, allowing the use of animal models to investigate mechanisms and pathogenesis. The molecular pathogenic mechanisms and comparative morphology in animals and humans will be reviewed.
Collapse
|
38
|
Gröschel C, Tennakoon S, Kállay E. Cytochrome P450 Vitamin D Hydroxylases in Inflammation and Cancer. ADVANCES IN PHARMACOLOGY 2015; 74:413-58. [PMID: 26233913 DOI: 10.1016/bs.apha.2015.03.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Vitamin D insufficiency correlates with increased incidence of inflammatory disorders and cancer of the colon, breast, liver, and prostate. Preclinical studies demonstrated that the hormonally active form of vitamin D, 1,25(OH)2D3, has antiproliferative, proapoptotic, anti-inflammatory, and immunomodulatory effects. Tissue levels of 1,25(OH)2D3 are determined by expression and activity of specific vitamin D hydroxylases expressed at renal and extrarenal sites. In order to understand how perturbations in the vitamin D system affect human health, we need to understand the steps involved in the synthesis and catabolism of the active metabolite. This review provides an overview about recent findings on the altered vitamin D metabolism in inflammatory conditions and carcinogenesis. We will summarize existing data on the pathophysiological regulation of vitamin D hydroxylases and outline the role of adequate levels of 1,25(OH)2D3 on tissue homeostasis.
Collapse
Affiliation(s)
- Charlotte Gröschel
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Währinger Gürtel, Vienna, Austria
| | - Samawansha Tennakoon
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Währinger Gürtel, Vienna, Austria
| | - Enikö Kállay
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Währinger Gürtel, Vienna, Austria.
| |
Collapse
|
39
|
Umbach AT, Zhang B, Daniel C, Fajol A, Velic A, Hosseinzadeh Z, Bhavsar SK, Bock CT, Kandolf R, Pichler BJ, Amann KU, Föller M, Lang F. Janus kinase 3 regulates renal 25-hydroxyvitamin D 1α-hydroxylase expression, calcitriol formation, and phosphate metabolism. Kidney Int 2014; 87:728-37. [PMID: 25493954 DOI: 10.1038/ki.2014.371] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 08/21/2014] [Accepted: 09/11/2014] [Indexed: 01/26/2023]
Abstract
Calcitriol, a powerful regulator of phosphate metabolism and immune response, is generated by 25-hydroxyvitamin D 1α-hydroxylase in the kidney and macrophages. Renal 1α-hydroxylase expression is suppressed by Klotho and FGF23, the expression of which is stimulated by calcitriol. Interferon γ (INFγ) regulates 1α-hydroxylase expression in macrophages through transcription factor interferon regulatory factor-1. INFγ-signaling includes Janus kinase 3 (JAK3) but a role of JAK3 in the regulation of 1α-hydroxylase expression and mineral metabolism has not been shown. Thus, the impact of JAK3 deficiency on calcitriol formation and phosphate metabolism was measured. Renal interferon regulatory factor-1 and 1α-hydroxylase transcript levels, serum calcitriol and FGF23 levels, intestinal phosphate absorption as well as absolute and fractional renal phosphate excretion were significantly higher in jak3 knockout than in wild-type mice. Coexpression of JAK3 increased the phosphate-induced current in renal sodium-phosphate cotransporter-expressing Xenopus oocytes. Thus, JAK3 is a powerful regulator of 1α-hydroxylase expression and phosphate transport. Its deficiency leads to marked derangement of phosphate metabolism.
Collapse
Affiliation(s)
- Anja T Umbach
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Bingbing Zhang
- 1] Department of Physiology, University of Tübingen, Tübingen, Germany [2] Department of Molecular Pathology, University of Tübingen, Tübingen, Germany
| | - Christoph Daniel
- Department of Nephropathology, University Hospital Erlangen, Erlangen, Germany
| | - Abul Fajol
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Ana Velic
- Proteome Center, University of Tübingen, Tübingen, Germany
| | | | - Shefalee K Bhavsar
- 1] Department of Physiology, University of Tübingen, Tübingen, Germany [2] Novartis Oncology, Novartis International AG, Hyderabad, India
| | - C-Thomas Bock
- Department of Molecular Pathology, University of Tübingen, Tübingen, Germany
| | - Reinhard Kandolf
- Department of Molecular Pathology, University of Tübingen, Tübingen, Germany
| | - Bernd J Pichler
- Department of Radiology, University of Tübingen, Tübingen, Germany
| | - Kerstin U Amann
- Department of Nephropathology, University Hospital Erlangen, Erlangen, Germany
| | - Michael Föller
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
40
|
Checkpoint kinase Chk2 controls renal Cyp27b1 expression, calcitriol formation, and calcium-phosphate metabolism. Pflugers Arch 2014; 467:1871-80. [PMID: 25319519 DOI: 10.1007/s00424-014-1625-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 09/29/2014] [Accepted: 10/01/2014] [Indexed: 02/08/2023]
Abstract
Checkpoint kinase 2 (Chk2) is the main effector kinase of ataxia telangiectasia mutated (ATM) and responsible for cell cycle regulation. ATM signaling has been shown to upregulate interferon-regulating factor-1 (IRF-1), a transcription factor also expressed in the kidney. Calcitriol (1,25 (OH)2D3), a major regulator of mineral metabolism, is generated by 25-hydroxyvitamin D 1α-hydroxylase in the kidney. Since 25-hydroxyvitamin D 1α-hydroxylase expression is enhanced by IRF-1, the present study explored the role of Chk2 for calcitriol formation and mineral metabolism. Chk2-deficient mice (chk2 (-/-)) were compared to wild-type mice (chk2 (+/+)). Transcript levels of renal 25-hydroxyvitamin D 1α-hydroxylase, Chk2, and IRF-1 were determined by RT-PCR; Klotho expression by Western blotting; bone density by μCT analysis; serum or plasma 1,25 (OH)2D3, PTH, and C-terminal FGF23 concentrations by immunoassays; and serum, fecal, and urinary calcium and phosphate concentrations by photometry. The renal expression of IRF-1 and 25-hydroxyvitamin D 1α-hydroxylase as well as serum 1,25 (OH)2D3 and FGF23 levels were significantly lower in chk2 (-/-) mice compared to chk2 (+/+) mice. Plasma PTH was not different between the genotypes. Renal calcium and phosphate excretion were significantly higher in chk2 (-/-) mice than in chk2 (+/+) mice despite hypophosphatemia and normocalcemia. Bone density was not different between the genotypes. We conclude that Chk2 regulates renal 25-hydroxyvitamin D 1α-hydroxylase expression thereby impacting on calcium and phosphate metabolism.
Collapse
|
41
|
Alshayeb H, Showkat A, Wall BM, Gyamlani GG, David V, Quarles LD. Activation of FGF-23 mediated vitamin D degradative pathways by cholecalciferol. J Clin Endocrinol Metab 2014; 99:E1830-7. [PMID: 24960544 PMCID: PMC4184071 DOI: 10.1210/jc.2014-1308] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CONTEXT The optimal circulating concentration of 25(OH) vitamin D is controversial. OBJECTIVE The aim was to investigate if FGF-23 and 24,25(OH)2D can guide cholecalciferol replacement. DESIGN Oral cholecalciferol (10,000 IU weekly) administered to subjects with 25(OH)D levels < 20 ηg/mL and eGFR > 60 mL/min/1.73 m(2) (n = 25), chronic kidney disease (CKD) (n = 27), or end stage renal disease (ESRD) (n = 14). SETTING The study was conducted at the Veterans Affairs clinics. MAIN OUTCOME MEASURE Serum FGF-23, PTH, 25(OH)D, 1,25(OH)2D, 24,25(OH)2D, calcium, and phosphorous concentrations, and urinary excretion of calcium and phosphorus at baseline and after 8 weeks of treatment. RESULTS Cholecalciferol treatment increased concentrations of serum 25(OH)D by (19.3 ± 8 ηg/mL, P = .001; 12.2 ± 9 ηg/mL, P = .0001) and 24,25(OH)2D (1.14 ± 0.89 ηg/mL, P = .0024; 1.0 ± 0.72 ηg/mL P = .0002), and reduced serum PTH (-11 ± 21 pg/mL, P = .0292; -42 ± 68 pg/mL, P = .0494) in normal and CKD subjects, respectively. Cholecalciferol increased serum FGF-23 levels only in normal subjects (44 ± 57 ηg/mL, P = .01). Increments in serum 25(OH)D positively correlated with serum FGF-23 and 24,25(OH)2D and negatively correlated with PTH. In ESRD, cholecalciferol administration increased 25(OH)D by (16.6 ± 6.6 ηg/mL P ≤ .05) without changing 24,25(OH)2D, FGF-23 or PTH levels. CONCLUSION Modest elevations of serum 25(OH)D levels after cholecalciferol treatment are sufficient to induce compensatory degradative pathways in patients with sufficient renal reserves, suggesting that optimal circulating 25(OH)D levels are approximately 20 ηg/mL. In addition, catabolism of 25(OH)D may also contribute to the low circulating vitamin D levels in CKD, since elevations of FGF-23 in CKD are associated with increased 24,25(OH)2D after cholecalciferol administration.
Collapse
Affiliation(s)
- Hala Alshayeb
- University of Tennessee Health Science Center (A.S., B.M.W., L.D.Q.), Memphis, Tennessee 38103; Veterans Affairs Medical Center (B.M.W., G.G.G.), Memphis, Tennessee 38103; Marshall University (H.A.), Huntington, West Virginia 25701; and University of Miami (V.D.), Miami, Florida 33136
| | | | | | | | | | | |
Collapse
|
42
|
Biber J, Murer H, Mohebbi N, Wagner C. Renal Handling of Phosphate and Sulfate. Compr Physiol 2014; 4:771-92. [DOI: 10.1002/cphy.c120031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
43
|
Block GA, Ix JH, Ketteler M, Martin KJ, Thadhani RI, Tonelli M, Wolf M, Jüppner H, Hruska K, Wheeler DC. Phosphate Homeostasis in CKD: Report of a Scientific Symposium Sponsored by the National Kidney Foundation. Am J Kidney Dis 2013; 62:457-73. [DOI: 10.1053/j.ajkd.2013.03.042] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 03/19/2013] [Indexed: 02/08/2023]
|
44
|
Abstract
Vitamin D metabolism consists of both production and catabolism, which are enzymatically driven and highly regulated. Renal vitamin D metabolism requires filtration and tubular reabsorption of 25-hydroxyvitamin D and is regulated by parathyroid hormone, fibroblast growth factor-23, and 1,25-dihydroxyvitamin D. In chronic kidney disease, renal production of 1,25-dihydroxyvitamin D from 25-hydroxyvitamin D is reduced. In addition, pharmacokinetic studies and epidemiologic studies of 24,25-dihydroxyvitamin D, the most abundant product of 25-hydroxyvitamin D catabolism by CYP24A1, suggest that vitamin D catabolism also is reduced. New insights into the mechanisms and regulation of vitamin D metabolism may lead to novel approaches to assess and treat impaired vitamin D metabolism in chronic kidney disease.
Collapse
Affiliation(s)
- Cortney Bosworth
- Division of Nephrology and Kidney Research Institute, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
45
|
Wang Z, Schuetz EG, Xu Y, Thummel KE. Interplay between vitamin D and the drug metabolizing enzyme CYP3A4. J Steroid Biochem Mol Biol 2013; 136:54-8. [PMID: 22985909 PMCID: PMC3549031 DOI: 10.1016/j.jsbmb.2012.09.012] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 09/04/2012] [Accepted: 09/07/2012] [Indexed: 12/19/2022]
Abstract
Cytochrome P450 3A4 (CYP3A4) is a multifunctional enzyme involved in both xenobiotic and endobiotic metabolism. This review focuses on two aspects: regulation of CYP3A4 expression by vitamin D and metabolism of vitamin D by CYP3A4. Enterohepatic circulation of vitamin D metabolites and their conjugates will be also discussed. The interplay between vitamin D and CYP3A4 provides new insights into our understanding of how enzyme induction can contribute to vitamin D deficiency. This article is part of a Special Issue entitled 'Vitamin D Workshop'.
Collapse
Affiliation(s)
- Zhican Wang
- Departments of Pharmaceutics, University of Washington, Seattle, WA
| | - Erin G. Schuetz
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - Yang Xu
- Departments of Pharmaceutics, University of Washington, Seattle, WA
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc, Thousand Oaks, CA
| | | |
Collapse
|
46
|
Abstract
Vitamin D is a dietary vitamin that can also be synthesized in adequate amounts from cholesterol in most mammals exposed to sunlight. Vitamin D has classical roles in calcium and phosphate metabolism, and thus the skeleton; however, this molecule also has nonclassical effects that might influence the function of the immune, cardiovascular and endocrine systems. Vitamin D deficiency, due to insufficient sunlight exposure, dietary uptake and/or abnormalities in its metabolism, has been associated with rheumatic diseases, and both the classical and nonclassical effects of vitamin D might be of relevance to patients with rheumatic disease. However, conclusive data from intervention trials demonstrating the relationship between vitamin D levels and pathogenetic processes separate from classical effects of this molecule are lacking. Furthermore, the majority of studies linking vitamin D to health outcomes, harmful or beneficial, are observational in nature, linking clinical events to vitamin D exposure or serum levels of vitamin D metabolites. Evidence from high quality, prospective, double-blind, placebo-controlled, randomized trials should be obtained before vitamin D supplementation is recommended in the treatment of the many rheumatic conditions in which deficiency of this compound has been implicated. Herein, we review the evidence for vitamin D supplementation in the management of patients with rheumatic diseases.
Collapse
|
47
|
Fujiwara M, Namba N, Ozono K, Arisaka O, Yokoya S. Treatment of Hypophosphatemic Rickets with Phosphate and Active Vitamin D in Japan: A Questionnaire-based Survey. Clin Pediatr Endocrinol 2013. [PMID: 23966755 PMCID: PMC3748281 DOI: 10.1297/cpe.22.9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Hereditary hypophosphatemic rickets represented
by X-linked hypophosphatemic rickets (XLH) is a rare disorder characterized by
hypophosphatemia, elevated alkaline phosphatase (ALP) and undermineralization of bone.
Active vitamin D and phosphate are administered to correct hypophosphatemia and elevation
of ALP. Overtreatment with phosphate leads to secondary hyperparathyroidism, and a large
dose of active vitamin D has a risk of hypercalciuria. To understand the situation
concerning treatment of patients with hereditary hypophosphatemic rickets in Japan, we
conducted a questionnaire survey of pediatric endocrinologists. Answers were obtained from
53 out of 68 hospitals where the pediatric endocrinologists worked. One hundred and
thirty-five patients were treated in 28 hospitals during November 2009 and May 2010; 126
patients suffered from hereditary hypophosphatemic rickets, and 9 had hypophosphatemia
caused by other miscellaneous reasons. The distribution of patient age was as follows: 27
(21%) were between 6 mo and 6 yr of age, 39 (31%) were between 6 and 12 yr of age, and 60
(48%) were more than 12 yr of age. Active vitamin D was given to 123 patients, and
phosphate was given to 106 patients. As for the dose of phosphorus, 37.2–58.1 mg/kg/d was
given divided into 2 to 6 aliquots. There were various control targets of treatment,
including serum phosphate, serum ALP, rachitic change, urinary Ca/Cr, parathyroid hormone
and growth. It is very important to avoid side effects of these treatments. No evidence is
available about the optimal dose of phosphate or number of administrations in the
treatment of patients with hypophosphatemic rickets. Although there is a recommendation
for clinical management of patients with hypophosphatemic rickets, we should set a
clinical guideline for it in Japan.
Collapse
Affiliation(s)
- Makoto Fujiwara
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Description of the recent advances on the regulation of phosphate metabolism, gene mutations, and new approaches to treatment in patients with hypophosphatemic rickets. RECENT FINDINGS Fibroblast growth factor 23 (FGF23) overproduction may be a primary cause of hypophosphatemic rickets. Inactivating mutations of phosphate-regulating gene with homologies to endopeptidases on the X chromosome, dentin matrix acidic phosphoprotein 1, and ectonucleotide pyrophosphatase/phosphodiesterase 1 are associated with X-linked hypophosphatemic rickets, autosomal recessive hypophosphatemic rickets 1, and autosomal recessive hypophosphatemic rickets 2, respectively. Activating mutations of FGF23 gene is the cause of autosomal dominant hypophosphatemic rickets. Iron deficiency may affect autosomal dominant hypophosphatemic rickets phenotype by regulating FGF23 production.Current treatment with activated vitamin D metabolites and oral inorganic phosphate salts may partially correct skeletal lesions and linear growth in patients with hypophosphatemic rickets. However, some patients have poor improvement by the current treatment. SUMMARY Identification of the causative mutation in patients with hypophosphatemic rickets may be useful to confirm the diagnosis and probably for prognosis. Inhibition of FGF23 overproduction by anti-FGF23 neutralizing antibodies could be a future approach for treatment of patients with FGF23-dependent hypophosphatemic rickets.
Collapse
Affiliation(s)
- Giampiero I Baroncelli
- Pediatric Unit I, Department of Obstetrics, Gynecology and Pediatrics, University-Hospital, Pisa, Italy.
| | | | | |
Collapse
|
49
|
Wang XZ, Zhang JJ, Wang Y, Xiang HY, . JH, . Z. Nephrotoxicity of Acetamiprid on Male Mice and the Rescue Role of Vitamin E. ACTA ACUST UNITED AC 2012. [DOI: 10.3923/javaa.2012.2721.2726] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
50
|
Pettifor JM, Thandrayen K. Hypophosphatemic rickets: unraveling the role of FGF23. Calcif Tissue Int 2012; 91:297-306. [PMID: 23001439 DOI: 10.1007/s00223-012-9651-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 09/06/2012] [Indexed: 12/29/2022]
Abstract
The classification of the various forms of hypophosphatemic rickets has been rationalized by the discovery of the central role that fibroblast growth factor 23 (FGF23) plays in the pathogenesis of a number of genetic and acquired forms of the disease. Although the details of the interaction of FGF23 with other osteoblast/osteocyte-derived proteins remain unclear at present, the measurement of circulating levels of FGF23 appears to be a useful biochemical test in determining the various causes of hypophosphatemic rickets. Furthermore, animal studies suggest that agents interfering in the action of FGF23 might play important roles in the future management of the FGF23-mediated forms of rickets. Phase 1 and phase 2 trials in humans with X-linked hypophosphatemic rickets are currently under way.
Collapse
Affiliation(s)
- John M Pettifor
- MRC/Wits Developmental Pathways for Health Research Unit, Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, 7 York Rd, Parktown, Johannesburg 2193, South Africa.
| | | |
Collapse
|