1
|
Wilson R, Mukherjee-Roy N, Gattineni J. The role of fibroblast growth factor 23 in regulation of phosphate balance. Pediatr Nephrol 2024; 39:3439-3451. [PMID: 38874635 DOI: 10.1007/s00467-024-06395-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 06/15/2024]
Abstract
Phosphate is essential for numerous biological processes, and serum levels are tightly regulated to accomplish these functions. The regulation of serum phosphate in a narrow physiological range is a well-orchestrated process and involves the gastrointestinal (GI) tract, bone, kidneys, and several hormones, namely, parathyroid hormone, fibroblast growth factor 23 (FGF23), and 1,25-dihydroxyvitamin D (1,25 Vitamin D). Although primarily synthesized in the bone, FGF23, an endocrine FGF, acts on the kidney to regulate phosphate and Vitamin D homeostasis by causing phosphaturia and reduced levels of 1,25 Vitamin D. Recent studies have highlighted the complex regulation of FGF23 including transcriptional and post-translational modification and kidney-bone cross talk. Understanding FGF23 biology has led to the identification of novel therapeutic agents to treat diseases that disrupt phosphate metabolism secondary to FGF23. The focus of this review is to provide an overview of phosphate homeostasis, FGF23 biology, and the role of FGF23 in phosphate balance.
Collapse
Affiliation(s)
| | - Neije Mukherjee-Roy
- Division of Pediatric Nephrology, Department of Pediatrics, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, USA
| | - Jyothsna Gattineni
- Division of Pediatric Nephrology, Department of Pediatrics, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, USA.
| |
Collapse
|
2
|
Molin A. Human genetic diseases of phosphate and pyrophosphate metabolism. Arch Pediatr 2024; 31:4S13-4S20. [PMID: 39343468 DOI: 10.1016/s0929-693x(24)00152-0] [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] [Indexed: 10/01/2024]
Abstract
In humans, physiological bone and tooth mineralization is a complex cell-mediated process. Prerequisites for proper mineralization include sufficient amounts of minerals (calcium and phosphate [Pi]) to initiate the formation and the growth of apatite crystals and adequate amounts of mineralization inhibitors, such as pyrophosphate (PPi), to prevent uncontrolled extraskeletal mineralization. In this review, we provide an overview of the genetics of human disorders of mineralization, focusing on Pi and PPi metabolism and transport diseases, as the Pi/PPi ratio is an important determinant of crystal production in vivo. Variants in genes implicated in the homeostasis of this ratio may lead to a systemic or local increased Pi/PPi ratio, either by increasing the Pi concentration or by decreasing the PPi concentration, resulting in ectopic calcifications; conversely, variants may lead to a decreased Pi/PPi ratio, resulting in defective mineralization. Owing to the implication of common pathways and, occasionally, to some extent of clinical overlap, an accurate diagnosis and understanding of the pathophysiology of these disorders may be challenging. However, precise molecular characterization of these conditions not only facilitates their diagnosis, but also helps to gather evidence regarding the pathophysiology and phenotype-genotype correlation to improve medical care and develop innovative therapeutics.
Collapse
Affiliation(s)
- Arnaud Molin
- Université de Caen Normandie, UFR Santé, CHU Caen Normandie, BIOTARGEN UR 7450, Centre de Référence Maladies rares du métabolisme du calcium et du phosphate, Service de Génétique, F-14000, Caen, France.
| |
Collapse
|
3
|
Sandez Penidez SH, De Moreno De Le Blanc A, Gerez CL, Rollán GC. Quinoa snack elaborated with Lactiplantibacillus plantarum CRL 1964 sourdough increases the mineral bioavailability in mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024. [PMID: 39132918 DOI: 10.1002/jsfa.13815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/14/2024] [Accepted: 07/21/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Consumption of pseudocereal-based foods decreased in phytate concentration can provide better nutrition concerning mineral bioavailability. This study aimed to evaluate the mineral bioavailability of quinoa sourdough-based snacks in a murine model. The mice were divided into five groups. One group was fed with basal snacks; three control groups received quinoa-based snacks made from non-fermented dough, dough without inoculum, and chemically acidified dough; and the test group (GF) received quinoa snacks elaborated from sourdough fermented by a phytase-positive strain, Lactiplantibacillus plantarum CRL 1964. Food intake, body weight, and mineral concentration in blood and organs (liver, kidney, and femur) were determined. RESULTS Food consumption increased during the feeding period and had the highest (16.2-24.5%) consumption in the GF group. Body weight also increased during the 6-weeks of trial. The GF group showed higher (6.0-10.2%) body weight compared with the other groups from the fifth week. The concentrations of iron, zinc, calcium, magnesium, and phosphorus in blood, iron and phosphorus in the liver, manganese and magnesium in the kidney, and calcium and phosphorus in the femur increased significantly (1.1-2.7-fold) in the GF group compared to the control groups. CONCLUSION The diet that includes quinoa snacks elaborated with sourdough fermented by phytase-positive strain L. plantarum CRL 1964 increased the concentrations of minerals in the blood, liver, kidney, and femur of mice, counteracting the antinutritional effects of phytate. This study demonstrates that the diminution in phytate content and the consequent biofortification in minerals are a suitable tool for producing novel foods. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
| | | | - Carla L Gerez
- Centro de Referencia para Lactobacilos (CERELA) - CONICET, San Miguel de Tucumán, Argentina
| | - Graciela C Rollán
- Centro de Referencia para Lactobacilos (CERELA) - CONICET, San Miguel de Tucumán, Argentina
| |
Collapse
|
4
|
Ratsma DMA, Muller M, Koedam M, van Leeuwen JPTM, Zillikens MC, van der Eerden BCJ. Organic phosphate but not inorganic phosphate regulates Fgf23 expression through MAPK and TGF-ꞵ signaling. iScience 2024; 27:109625. [PMID: 38883842 PMCID: PMC11178987 DOI: 10.1016/j.isci.2024.109625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/12/2024] [Accepted: 03/26/2024] [Indexed: 06/18/2024] Open
Abstract
One of the main regulators of phosphate homeostasis is fibroblast growth factor 23 (FGF23), secreted by osteocytes. The effects of organic versus inorganic dietary phosphate on this homeostasis are unclear. This study used MC3T3-E1 FGF23-producing cells to examine the transcriptomic responses to these phosphates. Most importantly, the expression and secretion of FGF23 were only increased in response to organic phosphate. Gene ontology terms related to a response to environmental change were only enriched in cells treated with organic phosphate while cells treated with inorganic phosphate were enriched for terms associated with regulation of cellular phosphate metabolism. Inhibition of MAPK signaling diminished the response of Fgf23 to organic phosphate, suggesting it activates FGF23. TGF-β signaling inhibition increased Fgf23 expression after the addition of organic phosphate, while the negative TGF-β regulator Skil decreased this response. In summary, the observed differential response of FGF23-producing to phosphate types may have consequences for phosphate homeostasis.
Collapse
Affiliation(s)
- Danielle M A Ratsma
- Laboratory for Calcium and Bone Metabolism and Erasmus MC Bone Centre, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Max Muller
- Laboratory for Calcium and Bone Metabolism and Erasmus MC Bone Centre, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marijke Koedam
- Laboratory for Calcium and Bone Metabolism and Erasmus MC Bone Centre, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Johannes P T M van Leeuwen
- Laboratory for Calcium and Bone Metabolism and Erasmus MC Bone Centre, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - M Carola Zillikens
- Laboratory for Calcium and Bone Metabolism and Erasmus MC Bone Centre, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Bram C J van der Eerden
- Laboratory for Calcium and Bone Metabolism and Erasmus MC Bone Centre, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
5
|
Ventrello SW, McMurry NR, Edwards NM, Bain LJ. Chronic arsenic exposure affects stromal cells and signaling in the small intestine in a sex-specific manner. Toxicol Sci 2024; 198:303-315. [PMID: 38310360 DOI: 10.1093/toxsci/kfae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2024] Open
Abstract
Arsenic is a toxicant that is ingested through drinking water and food, exposing nearly 140 million people to levels above the 10 ppb guideline concentration. Studies have shown that arsenic affects intestinal stem cells (ISCs), but the mechanisms by which arsenic alters the formation of adult cells in the small intestine are not well understood. Signals derived from intestinal stromal cells initiate and maintain differentiation. The goal of this study is to evaluate arsenic's effect on intestinal stromal cells, including PdgfrαLo trophocytes, located proximal to the ISCs, and PdgfrαHi telocytes, located proximal to the transit-amplifying region and up the villi. Adult Sox9tm2Crm-EGFP mice were exposed to 0, 33, and 100 ppb sodium arsenite in their drinking water for 13 weeks, and sections of duodenum were examined. Flow cytometry indicated that arsenic exposure dose-responsively reduced Sox9+ epithelial cells and trended toward increased Pdgfrα+ cells. The trophocyte marker, CD81, was reduced by 10-fold and 9.0-fold in the 100 ppb exposure group in male and female mice, respectively. Additionally, a significant 2.2- to 3.1-fold increase in PdgfrαLo expression was found in male mice in trophocytes and Igfbp5+ cells. PdgfrαHi protein expression, a telocyte marker, was more prevalent along the villus/crypt structure in females, whereas Gli1 expression (telocytes) was reduced in male mice exposed to arsenic. Principle coordinate analysis confirmed the sex-dependent response to arsenic exposure, with an increase in trophocyte and decrease in telocyte marker expression observed in male mice. These results imply that arsenic alters intestinal mesenchymal cells in a sex-dependent manner.
Collapse
Affiliation(s)
- Scott W Ventrello
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, USA
| | - Nicholas R McMurry
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, USA
| | - Nicholas M Edwards
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, USA
| | - Lisa J Bain
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, USA
| |
Collapse
|
6
|
Salcedo-Betancourt JD, Moe OW. The Effects of Acid on Calcium and Phosphate Metabolism. Int J Mol Sci 2024; 25:2081. [PMID: 38396761 PMCID: PMC10889523 DOI: 10.3390/ijms25042081] [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: 01/03/2024] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
A variety of changes in mineral metabolism aiming to restore acid-base balance occur in acid loading and metabolic acidosis. Phosphate plays a key role in defense against metabolic acidosis, both as an intracellular and extracellular buffer, as well as in the renal excretion of excess acid in the form of urinary titratable acid. The skeleton acts as an extracellular buffer in states of metabolic acidosis, as the bone matrix demineralizes, leading to bone apatite dissolution and the release of phosphate, calcium, carbonate, and citrate into the circulation. The renal handling of calcium, phosphate and citrate is also affected, with resultant hypercalciuria, hyperphosphaturia and hypocitraturia.
Collapse
Affiliation(s)
- Juan D. Salcedo-Betancourt
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Orson W. Moe
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
7
|
Alankarage D, Betts A, Scheckel KG, Herde C, Cavallaro M, Juhasz AL. Remediation options to reduce bioaccessible and bioavailable lead and arsenic at a smelter impacted site - consideration of treatment efficacy. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 341:122881. [PMID: 37935301 PMCID: PMC10843775 DOI: 10.1016/j.envpol.2023.122881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/30/2023] [Accepted: 11/04/2023] [Indexed: 11/09/2023]
Abstract
In this study, smelter contaminated soil was treated with various soil amendments (ferric sulfate [Fe2(SO4)3], triple superphosphate [TSP] and biochar) to determine their efficacy in immobilizing soil lead (Pb) and arsenic (As). In soils incubated with ferric sulfate (0.6M), gastric phase Pb bioaccessibility was reduced from 1939 ± 17 mg kg-1 to 245 ± 4.7 mg kg-1, while intestinal phase bioaccessibility was reduced from 194 ± 25 mg kg-1 to 11.9 ± 3.5 mg kg-1, driven by the formation of plumbojarosite. In TSP treated soils, there were minor reductions in gastric phase Pb bioaccessibility (to 1631 ± 14 mg kg-1) at the highest TSP concentration (6000 mg kg-1) although greater reductions were observed in the intestinal phase, with bioaccessibility reduced to 9.3 ± 2.2 mg kg-1. Speciation analysis showed that this was primarily driven by the formation of chloropyromorphite in the intestinal phase following Pb and phosphate solubilization in the low pH gastric fluid. At the highest concentration (10% w/w), biochar treated soils showed negligible decreases in Pb bioaccessibility in both gastric and intestinal phases. Validation of bioaccessibility outcomes using an in vivo mouse assay led to similar results, with treatment effect ratios (TER) of 0.20 ± 0.01, 0.76 ± 0.11 and 1.03 ± 0.10 for ferric sulfate (0.6M), TSP (6000 mg kg-1) and biochar (10% w/w) treatments. Results of in vitro and in vivo assays showed that only ferric sulfate treatments were able to significantly reduce As bioaccessibility and bioavailability with TER at the highest application of 0.06 ± 0.00 and 0.14 ± 0.04 respectively. This study highlights the potential application of ferric sulfate treatment for the immobilization of Pb and As in co-contaminated soils.
Collapse
Affiliation(s)
- Dileepa Alankarage
- Future Industries Institute, STEM, University of South Australia, SA, Australia.
| | - Aaron Betts
- United States Environmental Protection Agency, National Risk Management Research Laboratory, Land Remediation and Pollution Control Division, Cincinnati, OH, USA
| | - Kirk G Scheckel
- United States Environmental Protection Agency, National Risk Management Research Laboratory, Land Remediation and Pollution Control Division, Cincinnati, OH, USA
| | - Carina Herde
- South Australian Health and Medical Research Institute, Preclinical, Imaging and Research Laboratories, Adelaide, 5086, Australia
| | - Michelle Cavallaro
- South Australian Health and Medical Research Institute, Preclinical, Imaging and Research Laboratories, Adelaide, 5086, Australia
| | - Albert L Juhasz
- Future Industries Institute, STEM, University of South Australia, SA, Australia
| |
Collapse
|
8
|
MacDonald T, Beggs MR, O'Neill D, Kozuka K, Dimke H, Alexander RT. Increased Slc34a2 expression and paracellular phosphate permeability contribute to high intestinal phosphate absorption in young mice. Acta Physiol (Oxf) 2023; 239:e14029. [PMID: 37563989 DOI: 10.1111/apha.14029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023]
Abstract
AIM Phosphorus is a critical constituent of bone as a component of hydroxyapatite. Bone mineral content accrues rapidly early in life necessitating a positive phosphorus balance, which could be established by a combination of increased renal reabsorption and intestinal absorption. Intestinal absorption can occur via a transcellular pathway mediated by the apical sodium-phosphate cotransporter, Slc34a2/NaPiIIb or via the paracellular pathway. We sought to determine how young mammals increase dietary phosphorus absorption from the small intestine to establish a positive phosphorus balance, a prerequisite for rapid bone growth. METHODS The developmental expression profile of genes mediating phosphate absorption from the small intestine was determined in mice by qPCR and immunohistochemistry. Additionally, Ussing chamber studies were performed on small bowel of young (p7-p14) and older (8- to 17-week-old) mice to examine developmental changes in paracellular Pi permeability and transcellular Pi transport. RESULTS Blood and urinary Pi levels were higher in young mice. Intestinal paracellular phosphate permeability of young mice was significantly increased relative to older mice across all intestinal segments. NaPiIIb expression was markedly increased in juvenile mice, in comparison to adult animals. Consistent with this, young mice had increased transcellular phosphate flux across the jejunum and ileum relative to older animals. Moreover, transcellular phosphate transport was attenuated by the NaPiIIb inhibitor NTX1942 in the jejunum and ileum of young mice. CONCLUSION Our results are consistent with young mice increasing phosphate absorption via increasing paracellular permeability and the NaPiIIb-mediated transcellular pathway.
Collapse
Affiliation(s)
- Tate MacDonald
- Department of Physiology, The University of Alberta, Edmonton, Alberta, Canada
- The Women and Children's Health Research Institute, Edmonton, Alberta, Canada
| | - Megan R Beggs
- Department of Physiology, The University of Alberta, Edmonton, Alberta, Canada
- The Women and Children's Health Research Institute, Edmonton, Alberta, Canada
| | - Debbie O'Neill
- Department of Physiology, The University of Alberta, Edmonton, Alberta, Canada
| | | | - Henrik Dimke
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Nephrology, Odense University Hospital, Odense, Denmark
| | - R Todd Alexander
- Department of Physiology, The University of Alberta, Edmonton, Alberta, Canada
- The Women and Children's Health Research Institute, Edmonton, Alberta, Canada
- Department of Pediatrics, The University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
9
|
Wang H, Juhasz AL, Zhang Y, Zhang L, Ma LQ, Zhou D, Li H. Alcohol consumption promotes arsenic absorption but reduces tissue arsenic accumulation in mice. ECO-ENVIRONMENT & HEALTH (ONLINE) 2023; 2:107-116. [PMID: 38074988 PMCID: PMC10702898 DOI: 10.1016/j.eehl.2023.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/11/2023] [Accepted: 06/25/2023] [Indexed: 01/12/2024]
Abstract
Alcohol consumption alters gut microflora and damages intestinal tight junction barriers, which may affect arsenic (As) oral bioavailability. In this study, mice were exposed to arsenate in the diet (6 μg/g) over a 3-week period and gavaged daily with Chinese liquor (0.05 or 0.10 mL per mouse per day). Following ingestion, 78.0% and 72.9% of the total As intake was absorbed and excreted via urine when co-exposed with liquor at daily doses of 0.05 or 0.10 mL, significantly greater than when As was supplied alone (44.7%). Alcohol co-exposure significantly altered gut microbiota but did not significantly alter As biotransformation in the intestinal tract or tissue. Significantly lower relative mRNA expression was observed for genes encoding for tight junctions in the ileum of liquor co-exposed mice, contributing to greater As bioavailability attributable to enhanced As absorption via the intestinal paracellular pathway. However, As concentration in the liver, kidney, and intestinal tissue of liquor-treated mice was decreased by 24.4%-42.6%, 27.5%-38.1%, and 28.1%-48.9% compared to control mice. This was likely due to greater renal glomerular filtration rate induced by alcohol, as suggested by significantly lower expression of genes encoding for renal tight junctions. In addition, in mice gavaged daily with 0.05 mL liquor, the serum antidiuretic hormone level was significantly lower than control mice (2.83 ± 0.59 vs. 5.40 ± 1.10 pg/mL), suggesting the diuretic function of alcohol consumption, which may facilitate As elimination via urine. These results highlight that alcohol consumption has a significant impact on the bioavailability and accumulation of As.
Collapse
Affiliation(s)
- Hongyu Wang
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Albert L. Juhasz
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Yaosheng Zhang
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Lizhu Zhang
- Department of Nanxin Pharm, Nanjing 210000, China
| | - Lena Q. Ma
- Institute of Soil and Water Resources and Environmental Science, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Dongmei Zhou
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Hongbo Li
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| |
Collapse
|
10
|
Portales-Castillo I, Rieg T, Khalid SB, Nigwekar SU, Neyra JA. Physiopathology of Phosphate Disorders. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:177-188. [PMID: 36868732 PMCID: PMC10565570 DOI: 10.1053/j.akdh.2022.12.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/24/2022] [Accepted: 12/29/2022] [Indexed: 03/05/2023]
Abstract
Intracellular phosphate is critical for cellular processes such as signaling, nucleic acid synthesis, and membrane function. Extracellular phosphate (Pi) is an important component of the skeleton. Normal levels of serum phosphate are maintained by the coordinated actions of 1,25-dihydroxyvitamin D3, parathyroid hormone and fibroblast growth factor-23, which intersect in the proximal tubule to control the reabsorption of phosphate via the sodium-phosphate cotransporters Npt2a and Npt2c. Furthermore, 1,25-dihydroxyvitamin D3 participates in the regulation of dietary phosphate absorption in the small intestine. Clinical manifestations associated with abnormal serum phosphate levels are common and occur as a result of genetic or acquired conditions affecting phosphate homeostasis. For example, chronic hypophosphatemia leads to osteomalacia in adults and rickets in children. Acute severe hypophosphatemia can affect multiple organs leading to rhabdomyolysis, respiratory dysfunction, and hemolysis. Patients with impaired kidney function, such as those with advanced CKD, have high prevalence of hyperphosphatemia, with approximately two-thirds of patients on chronic hemodialysis in the United States having serum phosphate levels above the recommended goal of 5.5 mg/dL, a cutoff associated with excess risk of cardiovascular complications. Furthermore, patients with advanced kidney disease and hyperphosphatemia (>6.5 mg/dL) have almost one-third excess risk of death than those with phosphate levels between 2.4 and 6.5 mg/dL. Given the complex mechanisms that regulate phosphate levels, the interventions to treat the various diseases associated with hypophosphatemia or hyperphosphatemia rely on the understanding of the underlying pathobiological mechanisms governing each patient condition.
Collapse
Affiliation(s)
- Ignacio Portales-Castillo
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, and Harvard Medical School, Boston, MA; Endocrine Unit, Massachusetts General Hospital, and Harvard Medical School, Boston, MA
| | - Timo Rieg
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL; James A. Haley Veterans' Hospital, Tampa, FL; Center for Hypertension and Kidney Research, University of South Florida, Tampa, FL
| | - Sheikh B Khalid
- Department of Internal Medicine, The Indus Hospital, Lahore Pakistan
| | - Sagar U Nigwekar
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, and Harvard Medical School, Boston, MA
| | - Javier A Neyra
- Department of Internal Medicine, Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL.
| |
Collapse
|
11
|
Jones CLC, Shafer ABA, Frost PC. Characterizing nutritional phenotypes using experimental nutrigenomics: Is there nutrient-specificity to different types of dietary stress? Mol Ecol 2023; 32:1073-1086. [PMID: 36528862 DOI: 10.1111/mec.16825] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/05/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
The ability to directly measure and monitor poor nutrition in individual animals and ecological communities is hampered by methodological limitations. In this study, we use nutrigenomics to identify nutritional biomarkers in a freshwater zooplankter, Daphnia pulex, a ubiquitous primary consumer in lakes and a sentinel of environmental change. We grew animals in six ecologically relevant nutritional treatments: nutrient replete, low carbon (food), low phosphorus, low nitrogen, low calcium and high Cyanobacteria. We extracted RNA for transcriptome sequencing to identify genes that were nutrient responsive and capable of predicting nutritional status with a high degree of accuracy. We selected a list of 125 candidate genes, which were subsequently pruned to 13 predictive potential biomarkers. Using a nearest-neighbour classification algorithm, we demonstrate that these potential biomarkers are capable of classifying our samples into the correct nutritional group with 100% accuracy. The functional annotation of the selected biomarkers revealed some specific nutritional pathways and supported our hypothesis that animal responses to poor nutrition are nutrient specific and not simply different presentations of slow growth or energy limitation. This is a key step in uncovering the causes and consequences of nutritional limitation in animal consumers and their responses to small- and large-scale changes in biogeochemical cycles.
Collapse
Affiliation(s)
- Catriona L C Jones
- Environmental and Life Sciences Graduate Program, Trent University, Peterborough, Ontario, Canada
| | - Aaron B A Shafer
- Environmental and Life Sciences Graduate Program, Trent University, Peterborough, Ontario, Canada.,Department of Forensic Science, Trent University, Peterborough, Ontario, Canada
| | - Paul C Frost
- Department of Biology, Trent University, Peterborough, Ontario, Canada
| |
Collapse
|
12
|
Qi T, Wu L, Yu J, Song Z, Liu F, Li J, Song X, Li X. Acute low-dose phosphate disrupts glycerophospholipid metabolism and induces stress in juvenile turbot (Scophthalmus maximus). THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 861:160430. [PMID: 36455734 DOI: 10.1016/j.scitotenv.2022.160430] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/03/2022] [Accepted: 11/19/2022] [Indexed: 06/17/2023]
Abstract
Phosphate, as the main nutrient factor of lake eutrophication brought by pollutants discharged from agriculture and industry, is always considered to be a low-toxicity substance to aquatic animals. But the toxicity mechanism is unclear, and published information is limited. In this study, a 96 h acute stress experiment was conducted on juvenile turbot (Scophthalmus maximus) with 0, 10, and 60 mg/L phosphate solutions. Metabonomic analysis revealed that low-dose phosphate (10 mg/L) disrupted glycerophospholipid, purine, and glycolipid metabolism, as well as the tricarboxylic acid (TCA) cycle in juveniles, even at 96 h of stress, which may lead to cell structure damage and signal recognition disorder between cells. Upregulated key genes in the main glycerophospholipid metabolic pathways, which matched the results of the metabolomic study, were detected. Furthermore, low-dose phosphate (10 mg/L) induced oxidative stress and immunotoxicity in fish, resulting in the raising of relevant genes expression such as cat and sod in liver and kidney. In addition, all phosphate-treated groups had induced lesions on gill tissue, as evidenced by pathological observations. In this study on toxic effects on and mechanism of phosphate in aquatic animals using metabolomics, gene expression, and histopathology, we confirm that acute low-dose phosphate could disrupt glycerophospholipid metabolism and induce stress in juvenile turbot. This can provide advice on the amount of phosphate accumulation for marine fish farming and on protecting species diversity and marine ecosystem from the point of view of phosphate toxicity to marine animals.
Collapse
Affiliation(s)
- Ting Qi
- Key Laboratory of Mariculture (Ministry of Education), Fisheries College, Ocean University of China, Qingdao 266001, PR China
| | - Lele Wu
- Key Laboratory of Mariculture (Ministry of Education), Fisheries College, Ocean University of China, Qingdao 266001, PR China
| | - Jiachen Yu
- School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang, Jiangsu 222005, PR China
| | - Zongcheng Song
- Weihai Shenghang Aquatic Product Science and Technology Co. Ltd, Weihai 264200, PR China
| | - Feng Liu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China
| | - Jun Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China
| | - Xiefa Song
- Key Laboratory of Mariculture (Ministry of Education), Fisheries College, Ocean University of China, Qingdao 266001, PR China
| | - Xian Li
- Key Laboratory of Mariculture (Ministry of Education), Fisheries College, Ocean University of China, Qingdao 266001, PR China.
| |
Collapse
|
13
|
Wang HY, Chen S, Xue RY, Lin XY, Yang JL, Zhang YS, Li SW, Juhasz AL, Ma LQ, Zhou D, Li HB. Arsenic Ingested Early in Life Is More Readily Absorbed: Mechanistic Insights from Gut Microbiota, Gut Metabolites, and Intestinal Morphology and Functions. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:1017-1027. [PMID: 36580282 DOI: 10.1021/acs.est.2c04584] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Early-life arsenic (As) exposure is a particular health concern. However, it is unknown if As ingested early in life is more readily absorbed from the gastrointestinal (GI) tract, i.e., higher in oral bioavailability. Here, weanling (3-week) and adult (6-week-old) female mice were exposed to arsenate in the diet (10 μg g-1) over a 3-week period with As oral bioavailability estimated using As urinary excretion as the bioavailability endpoint. The As urinary excretion factor was 1.54-fold higher in weanling mice compared to adult mice (82.2 ± 7.29 versus 53.1 ± 3.73%), while weanling mice also showed 2.28-, 1.50-, 1.48-, and 1.89-fold higher As concentration in small intestine tissue, blood, liver, and kidneys, demonstrating significantly higher As oral bioavailability of early-life exposure. Compared to adult mice, weanling mice significantly differed in gut microbiota, but the difference did not lead to remarkable differences in As biotransformation in the GI tract or tissue and in overall gut metabolite composition. Although the expression of several metabolites (e.g., atrolactic acid, hydroxyphenyllactic acid, and xanthine) was up-regulated in weanling mice, they had limited ability to elevate As solubility in the intestinal tract. Compared to adult mice, the intestinal barrier function and intestinal expression of phosphate transporters responsible for arsenate absorption were similar in weanling mice. However, the small intestine of weanling mice was characterized by more defined intestinal villi with greater length and smaller width, providing a greater surface area for As to be absorbed across the GI barrier. The results highlight that early-life As exposure can be more readily absorbed, advancing the understanding of its health risk.
Collapse
Affiliation(s)
- Hong-Yu Wang
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Shan Chen
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Rong-Yue Xue
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Xin-Ying Lin
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Jin-Lei Yang
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Yao-Sheng Zhang
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Shi-Wei Li
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Albert L Juhasz
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Lena Q Ma
- Institute of Soil and Water Resources and Environmental Science, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Dongmei Zhou
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Hong-Bo Li
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| |
Collapse
|
14
|
Docio P, Llorente-Pelayo S, García-Unzueta MT, Lavin-Gómez BA, Puente N, Mateos F, Riancho-Zarrabeitia L, Gonzalez-Lamuño D, Riancho JA. Mild Hypophosphatemia-Associated Conditions in Children: The Need for a Comprehensive Approach. Int J Mol Sci 2022; 24:687. [PMID: 36614129 PMCID: PMC9820661 DOI: 10.3390/ijms24010687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
To better understand the causes of hypophosphatemia in children, we evaluated all serum phosphate tests performed in a tertiary hospital with unexpected but persistent temporary or isolated hypophosphatemia over an 18 year period. We collected 29,279 phosphate tests from 21,398 patients, of which 268 (1.2%) had at least one result showing hypophosphatemia. We found that endocrinopathies (n = 60), tumors (n = 10), and vitamin D deficiency (n = 3) were the medical conditions most commonly associated with mild hypophosphatemia, but in many patients the cause was unclear. Among patients with endocrinopathies, those with diabetes mellitus were found to have lower mean serum phosphate levels (mean 3.4 mg/dL) than those with short stature (3.7 mg/dL) or thyroid disorders (3.7 mg/dL). In addition, we found a correlation between glycemia and phosphatemia in patients with diabetes. However, despite the potential relevance of monitoring phosphate homeostasis and the underlying etiologic mechanisms, renal phosphate losses were estimated in less than 5% of patients with hypophosphatemia. In the pediatric age group, malignancies, hypovitaminosis D, and endocrine disorders, mostly diabetes, were the most common causes of hypophosphatemia. This real-world study also shows that hypophosphatemia is frequently neglected and inadequately evaluated by pediatricians, which emphasizes the need for more education and awareness about this condition to prevent its potentially deleterious consequences.
Collapse
Affiliation(s)
- Pablo Docio
- Servicio de Pediatría, Hospital U M Valdecilla, Instituto de Investigación Marqués de Valdecilla (IDIVAL), Universidad de Cantabria, 39011 Santander, Spain
| | - Sandra Llorente-Pelayo
- Servicio de Pediatría, Hospital U M Valdecilla, Instituto de Investigación Marqués de Valdecilla (IDIVAL), Universidad de Cantabria, 39011 Santander, Spain
| | - María Teresa García-Unzueta
- Servicio de Análisis Clínicos, Hospital U M Valdecilla, Universidad de Cantabria, IDIVAL, 39011 Santander, Spain
| | - Bernardo A. Lavin-Gómez
- Servicio de Análisis Clínicos, Hospital U M Valdecilla, Universidad de Cantabria, IDIVAL, 39011 Santander, Spain
| | - Nuria Puente
- Servicio de Medicina Interna, Hospital U M Valdecilla, Universidad de Cantabria, IDIVAL, 39011 Santander, Spain
| | - Fátima Mateos
- Servicio de Análisis Clínicos, Hospital Sierrallana, 39300 Torrelavega, Spain
| | | | - Domingo Gonzalez-Lamuño
- Servicio de Pediatría, Hospital U M Valdecilla, Instituto de Investigación Marqués de Valdecilla (IDIVAL), Universidad de Cantabria, 39011 Santander, Spain
| | - José A. Riancho
- Servicio de Medicina Interna, Hospital U M Valdecilla, Universidad de Cantabria, IDIVAL, 39011 Santander, Spain
| |
Collapse
|
15
|
Li HB, Xue RY, Chen XQ, Lin XY, Shi XX, Du HY, Yin NY, Cui YS, Li LN, Scheckel KG, Juhasz AL, Xue XM, Zhu YG, Ma LQ. Ca Minerals and Oral Bioavailability of Pb, Cd, and As from Indoor Dust in Mice: Mechanisms and Health Implications. ENVIRONMENTAL HEALTH PERSPECTIVES 2022; 130:127004. [PMID: 36541774 PMCID: PMC9769408 DOI: 10.1289/ehp11730] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/21/2022] [Accepted: 11/22/2022] [Indexed: 05/19/2023]
Abstract
BACKGROUND Elevating dietary calcium (Ca) intake can reduce metal(loid)oral bioavailability. However, the ability of a range of Ca minerals to reduce oral bioavailability of lead (Pb), cadmium (Cd), and arsenic (As) from indoor dust remains unclear. OBJECTIVES This study evaluated the ability of Ca minerals to reduce Pb, Cd, and As oral bioavailability from indoor dust and associated mechanisms. METHODS A mouse bioassay was conducted to assess Pb, Cd, and As relative bioavailability (RBA) in three indoor dust samples, which were amended into mouse chow without and with addition of CaHPO 4 , CaCO 3 , Ca gluconate, Ca lactate, Ca aspartate, and Ca citrate at 200 - 5,000 μ g / g Ca . The mRNA expression of Ca and phosphate (P) transporters involved in transcellular Pb, Cd and As transport in the duodenum of mice was quantified using real-time polymerase chain reaction. Serum 1,25-Dihydroxyvitamin D3 [1,25 ( OH ) 2 D 3 ], parathyroid hormone (PTH), and renal CYP27B1 activity controlling 1,25 ( OH ) 2 D 3 synthesis were measured using ELISA kits. Metal(loid) speciation in the feces of mice was characterized using X-ray absorption near-edge structure (XANES) spectroscopy. RESULTS In general, mice exposed to each of the Ca minerals exhibited lower Pb-, Cd-, and As-RBA for three dusts. However, RBAs with the different Ca minerals varied. Among minerals, mice fed dietary CaHPO 4 did not exhibit lower duodenal mRNA expression of Ca transporters but did have the lowest Pb and Cd oral bioavailability at the highest Ca concentration (5,000 μ g / g Ca ; 51%-95% and 52%-74% lower in comparison with the control). Lead phosphate precipitates (e.g., chloropyromorphite) were observed in feces of mice fed dietary CaHPO 4 . In comparison, mice fed organic Ca minerals (Ca gluconate, Ca lactate, Ca aspartate, and Ca citrate) had lower duodenal mRNA expression of Ca transporters, but Pb and Cd oral bioavailability was higher than in mice fed CaHPO 4 . In terms of As, mice fed Ca aspartate exhibited the lowest As oral bioavailability at the highest Ca concentration (5,000 μ g / g Ca ; 41%-72% lower) and the lowest duodenal expression of P transporter (88% lower). The presence of aspartate was not associated with higher As solubility in the intestine. DISCUSSION Our study used a mouse model of exposure to household dust with various concentrations and species of Ca to determine whether different Ca minerals can reduce bioavailability of Pb, Cd, and As in mice and elucidate the mechanism(s) involved. This study can contribute to the practical application of optimal Ca minerals to protect humans from Pb, Cd, and As coexposure in the environment. https://doi.org/10.1289/EHP11730.
Collapse
Affiliation(s)
- Hong-Bo Li
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing, China
| | - Rong-Yue Xue
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing, China
| | - Xiao-Qiang Chen
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing, China
| | - Xin-Ying Lin
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing, China
| | - Xiao-Xia Shi
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing, China
| | - Hai-Yan Du
- Nanjing Institute of Geography & Limnology, Chinese Academy of Sciences, Nanjing, China
| | - Nai-Yi Yin
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, China
| | - Yan-Shan Cui
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, China
| | - Li-Na Li
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, China
| | - Kirk G. Scheckel
- National Risk Management Research Laboratory, Land Remediation and Pollution Control Division, U.S. Environmental Protection Agency, Cincinnati, Ohio, USA
| | - Albert L. Juhasz
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia, Australia
| | - Xi-Mei Xue
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
| | - Yong-Guan Zhu
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
| | - Lena Q. Ma
- Institute of Soil and Water Resources and Environmental Science, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
16
|
Ibrahim HAA, Helmy E, Amin A, Mahmoud D. Biochemical and Anthropometric Nutritional Assessment in Children Infected with COVID-19: A Cross-sectional Study. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.9782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: Severe acute respiratory syndrome has led to a pandemic of coronavirus disease 2019 (COVID-19). Malnutrition either biochemically or anthropometrically is a well-known risk factor for COVID-19 and may be the vice versa
Objectives : To investigate the prevalence of malnutrition in children infected with COVID-19 through evaluating the nutritional biomarkers such as serum electrolytes, serum albumin and hemoglobin together with the anthropometric assessment.
Methods: A cross sectional study that was conducted at ElMatria Teaching Hospital for all children admitted with confirmed COVID-19 over a period of 6 months from 1st February 2021 to the end of July, 2021. Nutritional biochemical evaluation included serum electrolytes particularly the potassium and other nutritional biomarkers such as serum albumin and hemoglobin. Nutritional anthropometric evaluation depended on BMI (body mass index), the height/length, weight for length and weight for height..The prevalence of malnutrition esp. hypokalemia was the main outcome.
Results: Hypokalemia was present in 21.8% of the study participants . Other nutritional biomarkers were found as hyponatremia, hypocalcemia , hypophosphatemia, hypomagnesemia were detected in 49.1% , 38.2%,21.8% and 34.5% of the study subjects respectively. Anthropometric malnutrition was present in most of the enrolled children with COVID-19 in the study (65.5 % (n= 36) )through which overweight and obese children occupied a greater percentage.
Conclusion: Malnutrition either biochemically or anthropometrically could be linked to COVID-19 in children. COVID-19 could have negative outcomes on the nutritional status such as electrolytes disturbances. Both malnutrition and COVID-19 are considered synergistic associations
Keywords: Malnutrition. COVID-19. Children. Hypokalemia. Obesity
Collapse
|
17
|
Abstract
Phosphate homeostasis is dependent on the interaction and coordination of four main organ systems: thyroid/parathyroids, gastrointestinal tract, bone and kidneys, and three key hormonal regulators, 1,25-hydroxyvitamin D3, parathyroid hormone and FGF23 with its co- factor klotho. Phosphorus is a critical nutritional element for normal cellular function, but in excess can be toxic to tissues, particularly the vasculature. As phosphate, it also has an important interaction and inter-dependence with calcium and calcium homeostasis sharing some of the same controlling hormones, although this is not covered in our article. We have chosen to provide a current overview of phosphate homeostasis only, focusing on the role of two major organ systems, the gastrointestinal tract and kidneys, and their contribution to the control of phosphate balance. We describe in some detail the mechanisms of intestinal and renal phosphate transport, and compare and contrast their regulation. We also consider a significant example of phosphate imbalance, with phosphate retention, which is chronic kidney disease; why consequent hyperphosphatemia is important, and some of the newer means of managing it.
Collapse
Affiliation(s)
- Joanne Marks
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, United Kingdom
| | - Robert J Unwin
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, United Kingdom; Department of Renal Medicine, University College London, London, United Kingdom.
| |
Collapse
|
18
|
Phosphorus Absorption and Excretion in Hybrid Sturgeon (Huso dauricus♀ X Acipenser schrenckii♂) Intubated with Different Ca/P Ratios. FISHES 2022. [DOI: 10.3390/fishes7030138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
To study the effect of Ca/P ratio on the P and Ca absorption and excretion in hybrid sturgeon (Huso dauricus♀ X Acipenser schrenckii♂), five groups of fish were intubated with 100 mg P·kg−1 BW with the Ca/P ratios of 0:1, 0.25:1, 0.5:1, 1:1, and 2:1. Plasma P concentrations were significantly elevated at Ca/P ratios below 2:1, and the highest value was obtained at Ca/P ratio of 0.5:1. Plasma Ca content was significantly increased at the highest Ca/P ratio. Urine P excretion rate in the fish intubated with Ca/P ratio of 0.5:1 was significantly higher than that of the groups with Ca/P ratios of 0:1 and 2:1. The highest urea excretion rates were observed at Ca/P ratio of 0.5:1 and 1:1. The total P excretion at 48 h post intubation reached about 30 mg·kg−1 BW, which was recorded for the group with Ca/P ratio of 0.5:1. The present study showed that P absorption efficiency was improved in hybrid sturgeon at Ca/P ratio of 0.5:1, indicating that P inclusion level in sturgeon feed can be further optimized to reduce dietary P input and lower the excessive undigested P discharge into the rearing water.
Collapse
|
19
|
Zhai H, Adeola O, Liu J. Phosphorus nutrition of growing pigs. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2022; 9:127-137. [PMID: 35573097 PMCID: PMC9079227 DOI: 10.1016/j.aninu.2022.01.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/28/2021] [Accepted: 01/31/2022] [Indexed: 12/17/2022]
Abstract
Phosphorus (P) is an essential nutrient for diverse biological processes, which aggregate to the animal's requirement for P, and nutritionists strive to meet this requirement accurately. The P demand for a growing pig comprises requirements for maintenance and tissue deposition. The P in feed ingredients, however, must be digested and absorbed before its ultimate partition between the 2 aforementioned requirement components. Phosphorus from various sources could behave differently during digestion and absorption, which results in their disparate bioavailability for pigs. The system of standardized total tract digestibility reflects true total tract digestibility of P and feed ingredient effects on specific endogenous P loss with relative ease of implementation, and this system guarantees satisfactory additivity in digestible P among the ingredients in a diet-the foundation for diet formulation. The basal endogenous P loss, which is much easier to measure than the specific endogenous P loss, is considered as part of the pig's maintenance requirement. With this arrangement, a digestibility framework is established both for measuring the P-providing capacity of various feed ingredients and for describing the pig's P requirement. This framework entails basic understanding of the function, digestion, absorption, excretion, and homeostasis of P as support pillars. Understanding the workings of this framework enables potential integration of factors such as environment conditions and disease status in future P requirement models. The current review discusses dietary sources, digestion, absorption, bioavailability and requirement of P for growing pigs to understand the status quo, revealing the points of consensus as well as those of debate, and to encourage further investigation to provide more clarity.
Collapse
Affiliation(s)
- Hengxiao Zhai
- Southwest University of Science and Technology, Mianyang, China
- DSM China Animal Nutrition Research Center, Bazhou, China
| | - Olayiwola Adeola
- Department of Animal Sciences, Purdue University, West Lafayette, United States
| | - Jingbo Liu
- Southwest University of Science and Technology, Mianyang, China
| |
Collapse
|
20
|
Aligail K, Dave JA, Ross IL. Tumor-induced osteomalacia: a case report. J Med Case Rep 2022; 16:22. [PMID: 35016725 PMCID: PMC8752187 DOI: 10.1186/s13256-021-03220-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 12/03/2021] [Indexed: 11/30/2022] Open
Abstract
Background Tumor-induced osteomalacia is a rare, acquired paraneoplastic syndrome, including hypophosphatemia, high serum alkaline phosphatase, reduced active vitamin D, suboptimal bone mineral density, bone pain, fragility fractures, and muscle weakness. Case presentation We report a case of 74–year–old male of mixed ancestry with hypophosphatemia resistant to treatment despite optimal compliance, associated with profound reduction of bone mineral density and multiple nontraumatic fractures, including bilateral rib fractures, lower-thoracic (T11, T12) vertebrae, and two fractures involving the surgical and anatomical neck of the right humerus. We discuss an approach to identifying the underlying cause of hypophosphatemia associated with fragility fractures, and options for management of this rare condition. Conclusion Although rare, tumor-induced osteomalacia can be diagnosed if a logical stepwise approach is implemented. Surgery could be curative if the tumor is properly located and is resectable.
Collapse
Affiliation(s)
- Khalid Aligail
- Division of Endocrinology, Department of Medicine, Faculty of Health Sciences, Groote Schuur Hospital and University of Cape Town, J47-85 Old Main Building, Private Bag X3, Observatory, Cape Town, 7935, South Africa
| | - Joel A Dave
- Division of Endocrinology, Department of Medicine, Faculty of Health Sciences, Groote Schuur Hospital and University of Cape Town, J47-85 Old Main Building, Private Bag X3, Observatory, Cape Town, 7935, South Africa
| | - Ian Louis Ross
- Division of Endocrinology, Department of Medicine, Faculty of Health Sciences, Groote Schuur Hospital and University of Cape Town, J47-85 Old Main Building, Private Bag X3, Observatory, Cape Town, 7935, South Africa.
| |
Collapse
|
21
|
Wang R, He M, Kang Y. Hypophosphatemia at Admission is Associated with Increased Mortality in COVID-19 Patients. Int J Gen Med 2021; 14:5313-5322. [PMID: 34526806 PMCID: PMC8435477 DOI: 10.2147/ijgm.s319717] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 08/18/2021] [Indexed: 02/05/2023] Open
Abstract
Background Electrolyte disturbances are commonly observed in patients with coronavirus disease 2019 (COVID-19) and associated with outcome in these patients. Our study was designed to examine whether hypophosphatemia is associated with mortality in COVID-19 patients. Methods Patients diagnosed with COVID-19 and hospitalized in Renmin Hospital of Wuhan University between January 30 and February 24, 2020 were included in this study. Patients were divided into two groups, a hypophosphatemia group and a non-hypophosphatemia group, based on a serum phosphate level of 0.8 mmol/L. Logistic regression was performed to analyze the relationship between hypophosphatemia and mortality. A locally weighted scatterplot smoothing (LOWESS) curve was plotted to show the detailed association between mortality rate and serum phosphate level. A Kaplan–Meier survival curve was drawn to compare the difference in cumulative survival between the two groups. Results Hypophosphatemia at admission occurred in 33 patients, with an incidence of 7.6%. The hypophosphatemia group had a significantly higher incidence of respiratory failure (54.5% vs 32.6%, p=0.013) and mortality (57.6% vs 15.2%, p<0.001). Multivariate logistic regression indicated that age (OR=1.059, p<0.001), oxygen saturation (OR=0.733, p<0.001), white blood cells (OR=1.428, p<0.001), lymphocytes (OR=0.075, p<0.001) and hypophosphatemia (OR=3.636, p=0.015) were independently associated with mortality in the included patients. The hypophosphatemia group had significantly shorter survival than the non-hypophosphatemia group (p<0.001). Conclusion Hypophosphatemia at admission is associated with increased mortality in COVID-19 patients. More attention and medical care should be given to COVID-19 patients with hypophosphatemia at admission.
Collapse
Affiliation(s)
- Ruoran Wang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Min He
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, People's Republic of China.,COVID19 Medical Team (Hubei) of West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yan Kang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, People's Republic of China.,COVID19 Medical Team (Hubei) of West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
22
|
Yee J, Rosenbaum D, Jacobs JW, Sprague SM. Small Intestinal Phosphate Absorption: Novel Therapeutic Implications. Am J Nephrol 2021; 52:522-530. [PMID: 34515051 DOI: 10.1159/000518110] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/24/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND Chronic kidney disease (CKD) affects approximately 15% of adults in the USA. As CKD progresses, urinary phosphate excretion decreases and results in phosphate retention and, eventually, hyperphosphatemia. As hyperphosphatemia is associated with numerous adverse outcomes, including increased cardiovascular mortality, reduction in phosphorus concentrations is a guideline-recommended, established clinical practice. Dietary phosphate restriction, dialysis, and phosphate binders are currently the only options for phosphate management. However, many patients with hyperphosphatemia have phosphorus concentrations >5.5 mg/dL, despite treatment. SUMMARY This review pre-sents recent advances in the understanding of intestinal phosphate absorption and therapeutic implications. Dietary phosphate is absorbed in the intestine through two distinct pathways, paracellular absorption and transcellular transport. Recent evidence indicates that the paracellular route accounts for 65-80% of total phosphate absorbed. Thus, the paracellular pathway is the dominant mechanism of phosphate absorption. Tenapanor is a first-in-class, non-phosphate binder that inhibits the sodium-hydrogen exchanger 3 or solute carrier family 9 member 3 (SLC9A3) encoded by the SLC9A3 gene, and blocks paracellular phosphate absorption. Key Messages: Targeted inhibition of sodium-hydrogen exchanger 3 effectively reduces paracellular permeability of phosphate. Novel therapies that target the paracellular pathway may improve phosphate control in chronic kidney disease.
Collapse
Affiliation(s)
- Jerry Yee
- Nephrology and Hypertension, Henry Ford Hospital, Detroit, Michigan, USA
| | | | | | - Stuart M Sprague
- Division of Nephrology and Hypertension, NorthShore University Health System, Evanston, Illinois, USA
| |
Collapse
|
23
|
Asowata EO, Olusanya O, Abaakil K, Chichger H, Srai SKS, Unwin RJ, Marks J. Diet-induced iron deficiency in rats impacts small intestinal calcium and phosphate absorption. Acta Physiol (Oxf) 2021; 232:e13650. [PMID: 33749990 DOI: 10.1111/apha.13650] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 02/26/2021] [Accepted: 03/17/2021] [Indexed: 12/31/2022]
Abstract
AIMS Recent reports suggest that iron deficiency impacts both intestinal calcium and phosphate absorption, although the exact transport pathways and intestinal segment responsible have not been determined. Therefore, we aimed to systematically investigate the impact of iron deficiency on the cellular mechanisms of transcellular and paracellular calcium and phosphate transport in different regions of the rat small intestine. METHODS Adult, male Sprague-Dawley rats were maintained on a control or iron-deficient diet for 2 weeks and changes in intestinal calcium and phosphate uptake were determined using the in situ intestinal loop technique. The circulating levels of the hormonal regulators of calcium and phosphate were determined by ELISA, while the expression of transcellular calcium and phosphate transporters, and intestinal claudins were determined using qPCR and western blotting. RESULTS Diet-induced iron deficiency significantly increased calcium absorption in the duodenum but had no impact in the jejunum and ileum. In contrast, phosphate absorption was significantly inhibited in the duodenum and to a lesser extent the jejunum, but remained unchanged in the ileum. The changes in duodenal calcium and phosphate absorption in the iron-deficient animals were associated with increased claudin 2 and 3 mRNA and protein levels, while levels of parathyroid hormone, fibroblast growth factor-23 and 1,25-dihydroxy vitamin D3 were unchanged. CONCLUSION We propose that iron deficiency alters calcium and phosphate transport in the duodenum. This occurs via changes to the paracellular pathway, whereby upregulation of claudin 2 increases calcium absorption and upregulation of claudin 3 inhibits phosphate absorption.
Collapse
Affiliation(s)
- Evans O. Asowata
- Department of Neuroscience, Physiology & Pharmacology University College London London UK
| | - Oluwatobi Olusanya
- Department of Neuroscience, Physiology & Pharmacology University College London London UK
| | - Kaoutar Abaakil
- Department of Neuroscience, Physiology & Pharmacology University College London London UK
| | - Havovi Chichger
- Biomedical Research Group School of Life Sciences Anglia Ruskin University Cambridge UK
| | - Surjit K. S. Srai
- Institute of Structural and Molecular Biology University College London London UK
| | - Robert J. Unwin
- Department of Renal Medicine University College London London UK
| | - Joanne Marks
- Department of Neuroscience, Physiology & Pharmacology University College London London UK
| |
Collapse
|
24
|
Chande S, Dijk F, Fetene J, Yannicelli S, Carpenter TO, van Helvoort A, Bergwitz C. Phosphorus bioaccessibility measured in four amino acid-based formulas using in-vitro batch digestion translates well into phosphorus bioavailability in mice. Nutrition 2021; 89:111291. [PMID: 34111672 DOI: 10.1016/j.nut.2021.111291] [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: 10/18/2020] [Revised: 04/07/2021] [Accepted: 04/18/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE The aim of this study was to quantify the bioaccessibility of phosphorus from amino acid-based formulas (AAFs) under different digestive conditions. METHODS We developed in-vitro batch digestion models with stomach digestion at different pH mimicking the normal digestive condition and conditions representing use of acid-suppressive medication. To validate bioaccessibility findings, we devised a low phosphorus murine model to test phosphorus bioavailability under compromised digestive conditions using proton pump inhibitors (PPIs) to neutralize stomach pH. RESULTS In vitro phosphorus bioaccessibility of AAFs Neocate® Infant and Neocate Junior ranged between 57% and 65% under normal digestive conditions for infants (stomach pH 3.5) and between 38% and 46% under conditions that simulated bypass of stomach acidification, which is comparable to control diet and two EleCare® AAFs. In vivo bioavailability analysis showed that both Neocate formulas were able to normalize plasma phosphorus levels when administered to low phosphorus mice along with PPIs (control diet + PPI 8 ± 0.4; Neocate Infant 10.1 ± 0.9; Neocate Junior 9.2 ± 0.6; EleCare Infant 8.6 ± 0.4; EleCare Junior 8.7 ± 0.5; n = 8-10; P < 0.0001 versus baseline 3.4 ± 0.2 mg/dL). In comparison, plasma phosphorus levels remained lower on the low phosphorus diet (5.7 ± 0.2 mg/dL). Furthermore, urinary phosphorus/creatinine and intact fibroblast growth factor 23 were significantly lowered by low phosphorus diet. In contrast, intact parathyroid hormone and 1,25-dihydroxy vitamin D decreased and increased, respectively, and these parameters likewise normalized in mice administered AAFs. CONCLUSION The present findings indicated that phosphorus bioaccessibility in the in-vitro batch digestion model translates well into phosphorus bioavailability in mice even under compromised digestive conditions that bypass gastric acidification.
Collapse
Affiliation(s)
- Sampada Chande
- Yale University School of Medicine, Section of Endocrinology and Metabolism, New Haven, Connecticut, USA
| | | | - Jonathan Fetene
- Yale University School of Medicine, Section of Endocrinology and Metabolism, New Haven, Connecticut, USA
| | | | - Thomas O Carpenter
- Yale University School of Medicine, Department of Pediatrics, New Haven, Connecticut, USA
| | - Ardy van Helvoort
- Danone Nutricia Research, Utrecht, The Netherlands; School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Clemens Bergwitz
- Yale University School of Medicine, Section of Endocrinology and Metabolism, New Haven, Connecticut, USA.
| |
Collapse
|
25
|
Ketteler M, Wiecek A, Rosenkranz AR, Pasch A, Rekowski J, Hellmann B, Karus M, Ammer R. Efficacy and Safety of a Novel Nicotinamide Modified-Release Formulation in the Treatment of Refractory Hyperphosphatemia in Patients Receiving Hemodialysis-A Randomized Clinical Trial. Kidney Int Rep 2021; 6:594-604. [PMID: 33732974 PMCID: PMC7938065 DOI: 10.1016/j.ekir.2020.12.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/08/2020] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Despite widespread use of phosphate binders (PBs), phosphate control is insufficient in many hemodialysis patients. Preliminary clinical observations suggest that nicotinamide may act synergistically with PBs to improve phosphate control. METHODS This multinational, randomized, double-blind, placebo-controlled study evaluated the efficacy and safety of nicotinamide modified release (NAMR) in combination with oral PB in a large cohort of hemodialysis patients with abnormal serum phosphate concentration (>4.5 mg/dl) despite treatment with PB. Patients entered a proof-of-efficacy phase (12 weeks [W12]) in which adjustments of relevant comedication were not permitted, followed by a safety extension phase for up to 52 weeks. Here, we report the results of the first phase. RESULTS The intention-to-treat (ITT) population consisted of 539 patients in the NAMR and 183 patients in the placebo group. NAMR and placebo were orally administered once daily (250-1500 mg/d). Mean age of patients was 61.8 years, and 63.0% were men. In the confirmatory analysis that estimated the difference in serum phosphate concentration after 12 weeks, NAMR proved superior over placebo with a significant difference of -0.51 mg/dl (95% confidence interval [CI] -0.72, -0.29; P < 0.0001). This effect was associated with significantly lower intact parathyroid hormone (iPTH) values (NAMR: 292.4±300.4 pg/ml vs. placebo: 337.0±302.7 pg/ml; P = 0.04) and an improved calcification propensity (T50 time; NAMR: 23.8±97.1 minutes vs. placebo: 2.3±100.7 minutes; P = 0.02). Diarrhea and pruritus were more frequent in the NAMR group. CONCLUSION NAMR combined with oral PB significantly improved phosphate control in hemodialysis patients.
Collapse
Affiliation(s)
- Markus Ketteler
- Robert Bosch Hospital, Department of General Internal Medicine and Nephrology, Stuttgart, Germany
- University of Split, School of Medicine, Split, Croatia
| | - Andrzej Wiecek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia in Katowice, Poland
| | - Alexander R. Rosenkranz
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Andreas Pasch
- Calciscon AG, Nidau, Switzerland
- Institute of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
- Nierenpraxis Bern, Bern, Switzerland
- Department of Nephrology, Lindenhofspital, Bern, Switzerland
| | - Jan Rekowski
- Institute for Medical Informatics, Biometry and Epidemiology, Essen University Hospital, Essen, Germany
- Center for Clinical Studies, University Hospital Essen, Essen, Germany
| | - Burkhard Hellmann
- Medical Department, MEDICE Arzneimittel Pütter GmbH & Co KG, Iserlohn, Germany
| | - Michael Karus
- Medical Department, MEDICE Arzneimittel Pütter GmbH & Co KG, Iserlohn, Germany
| | - Richard Ammer
- Medical Department, MEDICE Arzneimittel Pütter GmbH & Co KG, Iserlohn, Germany
- Department of Nephrology, University Hospital Münster, Münster, Germany
| |
Collapse
|
26
|
Verna G, Sila A, Liso M, Mastronardi M, Chieppa M, Cena H, Campiglia P. Iron-Enriched Nutritional Supplements for the 2030 Pharmacy Shelves. Nutrients 2021; 13:378. [PMID: 33530485 PMCID: PMC7912282 DOI: 10.3390/nu13020378] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/18/2021] [Accepted: 01/22/2021] [Indexed: 12/18/2022] Open
Abstract
Iron deficiency (ID) affects people of all ages in many countries. Due to intestinal blood loss and reduced iron absorption, ID is a threat to IBD patients, women, and children the most. Current therapies can efficiently recover normal serum transferrin saturation and hemoglobin concentration but may cause several side effects, including intestinal inflammation. ID patients may benefit from innovative nutritional supplements that may satisfy iron needs without side effects. There is a growing interest in new iron-rich superfoods, like algae and mushrooms, which combine antioxidant and anti-inflammatory properties with iron richness.
Collapse
Affiliation(s)
- Giulio Verna
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Annamaria Sila
- National Institute of Gastroenterology “S. de Bellis”, Institute of Research, 70013 Castellana Grotte, Italy; (A.S.); (M.L.); (M.M.); (M.C.)
| | - Marina Liso
- National Institute of Gastroenterology “S. de Bellis”, Institute of Research, 70013 Castellana Grotte, Italy; (A.S.); (M.L.); (M.M.); (M.C.)
| | - Mauro Mastronardi
- National Institute of Gastroenterology “S. de Bellis”, Institute of Research, 70013 Castellana Grotte, Italy; (A.S.); (M.L.); (M.M.); (M.C.)
| | - Marcello Chieppa
- National Institute of Gastroenterology “S. de Bellis”, Institute of Research, 70013 Castellana Grotte, Italy; (A.S.); (M.L.); (M.M.); (M.C.)
| | - Hellas Cena
- Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy;
- Clinical Nutrition and Dietetics Service, Unit of Internal Medicine and Endocrinology, ICS Maugeri I.R.C.C.S, 27100 Pavia, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| |
Collapse
|
27
|
Sellars KB, Ryan BA, Hartery SA, Kirby BJ, Kovacs CS. Murine Fetal Serum Phosphorus is Set Independent of FGF23 and PTH, Except in the Presence of Maternal Phosphate Loading. Endocrinology 2021; 162:5956315. [PMID: 33150413 PMCID: PMC7737482 DOI: 10.1210/endocr/bqaa202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Indexed: 01/03/2023]
Abstract
Fibroblast growth factor 23 (FGF23) appears to play no role until after birth, given unaltered phosphate and bone metabolism in Fgf23- and Klotho-null fetuses. However, in those studies maternal serum phosphorus was normal. We studied whether maternal phosphate loading alters fetal serum phosphorus and invokes a fetal FGF23 or parathyroid hormone (PTH) response. C57BL/6 wild-type (WT) female mice received low (0.3%), normal (0.7%), or high (1.65%) phosphate diets beginning 1 week prior to mating to WT males. Fgf23+/- female mice received the normal or high-phosphate diets 1 week before mating to Fgf23+/- males. One day before expected birth, we harvested maternal and fetal blood, intact fetuses, placentas, and fetal kidneys. Increasing phosphate intake in WT resulted in progressively higher maternal serum phosphorus and FGF23 during pregnancy, while PTH remained undetectable. Fetal serum phosphorus was independent of the maternal phosphorus and PTH remained low, but FGF23 showed a small nonsignificant increase with high maternal serum phosphorus. There were no differences in fetal ash weight and mineral content, or placental gene expression. High phosphate intake in Fgf23+/- mice also increased maternal serum phosphorus and FGF23, but there was no change in PTH. WT fetuses remained unaffected by maternal high-phosphate intake, while Fgf23-null fetuses became hyperphosphatemic but had no change in PTH, skeletal ash weight or mineral content. In conclusion, fetal phosphate metabolism is generally regulated independently of maternal serum phosphorus and fetal FGF23 or PTH. However, maternal phosphate loading reveals that fetal FGF23 can defend against the development of fetal hyperphosphatemia.
Collapse
Affiliation(s)
- K Berit Sellars
- Faculty of Medicine – Endocrinology, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Brittany A Ryan
- Faculty of Medicine – Endocrinology, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Sarah A Hartery
- Faculty of Medicine – Endocrinology, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Beth J Kirby
- Faculty of Medicine – Endocrinology, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Christopher S Kovacs
- Faculty of Medicine – Endocrinology, Memorial University of Newfoundland, St. John’s, NL, Canada
- Correspondence: Dr Christopher Kovacs, Health Sciences Centre, 300 Prince Philip Drive, St. John’s, Newfoundland, A1B 3V6, Canada. E-mail:
| |
Collapse
|
28
|
Abstract
Phosphorus, a 5A element with atomic weight of 31, comprises just over 0.6% of the composition by weight of plants and animals. Three isotopes are available for studying phosphorus metabolism and kinetics. 31P is stable, whereas the radioactive isotope 33P has a half-life of 25 days and 32P has a half-life of 14 days. Phosphate ester and phosphoanhydride are common chemical linkages and phosphorus is a key element in organic molecules involved in a wide variety of essential cellular functions. These include biochemical energy transfer via adenosine triphosphate (ATP), maintenance of genetic information with nucleotides DNA and RNA, intracellular signaling via cyclic adenosine monophosphate (cAMP), and membrane structural integrity via glycerophospholipids. However, this review focuses on the metabolism of inorganic phosphorus (Pi) acting as a weak acid. Phosphoric acid has all three hydrogens attached to oxygen and is a weak diprotic acid. It has 3 pKa values: pH 2.2, pH 7.2, and pH 12.7. At physiological pH of 7.4, Pi exists as both H2PO4(-) and HPO4(2-) and acts as an extracellular fluid (ECF) buffer. Pi is the form transported across tissue compartments and cells. Measurement of Pi in biological fluids is based on its reaction with ammonium molybdate which does not measure organic phosphorus. In humans, 80% of the body phosphorus is present in the form of calcium phosphate crystals (apatite) that confer hardness to bone and teeth, and function as the major phosphorus reservoir (Fig. 1). The remainder is present in soft tissues and ECF. Dietary phosphorus, comprising both inorganic and organic forms, is digested in the upper gastrointestinal tract. Absorbed Pi is transported to and from bone, skeletal muscle and soft tissues, and kidney at rates determined by ECF Pi concentration, rate of blood flow, and activity of cell Pi transporters (Fig. 2). During growth, there is net accretion of phosphorus, and with aging, net loss of phosphorus occurs. The bone phosphorus reservoir is depleted and repleted by overall phosphorus requirement. Skeletal muscle is rich in phosphorus used in essential biochemical energy transfer. Kidney is the main regulator of ECF Pi concentration by virtue of having a tubular maximum reabsorptive capacity for Pi (TmPi) that is under close endocrine control. It is also the main excretory pathway for Pi surplus which is passed in urine. Transcellular and paracellular Pi transports are performed by a number of transport mechanisms widely distributed in tissues, and particularly important in gut, bone, and kidney. Pi transporters are regulated by a hormonal axis comprising fibroblast growth factor 23 (FGF23), parathyroid hormone (PTH), and 1,25 dihydroxy vitamin D (1,25D). Pi and calcium (Ca) metabolism are intimately interrelated, and clinically neither can be considered in isolation. Diseases of Pi metabolism affect bone as osteomalacia/rickets, soft tissues as ectopic mineralization, skeletal muscle as myopathy, and kidney as nephrocalcinosis and urinary stone formation. Fig. 1 Content of phosphorus in human adult: skeleton, soft tissue, and extracellular fluid (grams, log scale). Corresponding data for calcium are shown for comparison Fig. 2 Phosphate (Pi) transport to and from tissue compartments in mg/24 h. At a dietary phosphorus of 1400 mg, 1120 mg is absorbed in upper intestine to the ECF, 210 mg returned to intestine by endogenous secretion, resulting in 910 mg net Pi absorption and 490 mg fecal excretion. At bone, 180 mg is deposited by bone formation and 180 mg return to the ECF by bone resorption. At kidney, 5040 mg is filtered at the glomerulus and 4130 mg return to the ECF by tubular reabsorption with 910 mg excreted in the urine. In soft tissue, Pi is exchanged between ECF and cells.
Collapse
Affiliation(s)
- Munro Peacock
- Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, 1120 W Michigan Street, CL365, Indianapolis, IN, 46202, USA.
| |
Collapse
|
29
|
Laflamme D, Backus R, Brown S, Butterwick R, Czarnecki-Maulden G, Elliott J, Fascetti A, Polzin D. A review of phosphorus homeostasis and the impact of different types and amounts of dietary phosphate on metabolism and renal health in cats. J Vet Intern Med 2020; 34:2187-2196. [PMID: 33159358 PMCID: PMC7694857 DOI: 10.1111/jvim.15961] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 10/11/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022] Open
Abstract
Elevated concentrations of serum phosphate are linked with progression and increased case fatality rate in animals and humans with chronic kidney disease. Elevated concentrations of serum phosphate can be a risk factor for development of renal and cardiovascular diseases or osteoporosis in previously healthy people. In rodents, an excess intake of dietary phosphorus combined with an inverse dietary calcium : phosphorus ratio (<1 : 1) contributes to renal calcification. Renal injury also has occured in cats fed experimental diets supplemented with highly soluble phosphate salts, especially in diets with inverse calcium : phosphorus ratios. However, not all phosphorus sources contribute similarly to this effect. This review, which focuses on cats, summarizes the published evidence regarding phosphorus metabolism and homeostasis, including the relative impact of different dietary phosphorus sources, and their impact on the kidneys. No data currently shows that commercial cat foods induce renal injury. However, some diets contain high amounts of phosphorus relative to recommendations and some have inverse Ca : P ratios and so could increase the risk for development of kidney disease. While limiting the use of highly soluble phosphates appears to be important, there are insufficient data to support a specific upper limit for phosphate intake. This review also proposes areas where additional research is needed in order to strengthen conclusions and recommendations regarding dietary phosphorus for cats.
Collapse
Affiliation(s)
| | - Robert Backus
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Scott Brown
- College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | | | | | - Jonathan Elliott
- Royal Veterinary College, University of London, London, United Kingdom
| | - Andrea Fascetti
- Department of Molecular Biosciences, University of California, Davis, California, USA
| | - David Polzin
- Veterinary Clinical Sciences Department, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota, USA
| |
Collapse
|
30
|
Serna J, Bergwitz C. Importance of Dietary Phosphorus for Bone Metabolism and Healthy Aging. Nutrients 2020; 12:E3001. [PMID: 33007883 PMCID: PMC7599912 DOI: 10.3390/nu12103001] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 12/13/2022] Open
Abstract
Inorganic phosphate (Pi) plays a critical function in many tissues of the body: for example, as part of the hydroxyapatite in the skeleton and as a substrate for ATP synthesis. Pi is the main source of dietary phosphorus. Reduced bioavailability of Pi or excessive losses in the urine causes rickets and osteomalacia. While critical for health in normal amounts, dietary phosphorus is plentiful in the Western diet and is often added to foods as a preservative. This abundance of phosphorus may reduce longevity due to metabolic changes and tissue calcifications. In this review, we examine how dietary phosphorus is absorbed in the gut, current knowledge about Pi sensing, and endocrine regulation of Pi levels. Moreover, we also examine the roles of Pi in different tissues, the consequences of low and high dietary phosphorus in these tissues, and the implications for healthy aging.
Collapse
Affiliation(s)
- Juan Serna
- Yale College, Yale University, New Haven, CT 06511, USA;
| | - Clemens Bergwitz
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06519, USA
| |
Collapse
|
31
|
Xie Y, Su N, Yang J, Tan Q, Huang S, Jin M, Ni Z, Zhang B, Zhang D, Luo F, Chen H, Sun X, Feng JQ, Qi H, Chen L. FGF/FGFR signaling in health and disease. Signal Transduct Target Ther 2020; 5:181. [PMID: 32879300 PMCID: PMC7468161 DOI: 10.1038/s41392-020-00222-7] [Citation(s) in RCA: 379] [Impact Index Per Article: 94.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/28/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022] Open
Abstract
Growing evidences suggest that the fibroblast growth factor/FGF receptor (FGF/FGFR) signaling has crucial roles in a multitude of processes during embryonic development and adult homeostasis by regulating cellular lineage commitment, differentiation, proliferation, and apoptosis of various types of cells. In this review, we provide a comprehensive overview of the current understanding of FGF signaling and its roles in organ development, injury repair, and the pathophysiology of spectrum of diseases, which is a consequence of FGF signaling dysregulation, including cancers and chronic kidney disease (CKD). In this context, the agonists and antagonists for FGF-FGFRs might have therapeutic benefits in multiple systems.
Collapse
Affiliation(s)
- Yangli Xie
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| | - Nan Su
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jing Yang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Qiaoyan Tan
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Shuo Huang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Min Jin
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhenhong Ni
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Bin Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Dali Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Fengtao Luo
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Hangang Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xianding Sun
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Huabing Qi
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
32
|
Kaesler N, Goettsch C, Weis D, Schurgers L, Hellmann B, Floege J, Kramann R. Magnesium but not nicotinamide prevents vascular calcification in experimental uraemia. Nephrol Dial Transplant 2020; 35:65-73. [PMID: 30715488 DOI: 10.1093/ndt/gfy410] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 12/13/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Optimal phosphate control is an unmet need in chronic kidney disease (CKD). High serum phosphate increases calcification burden and is associated with mortality and cardiovascular disease in CKD. Nicotinamide (NA) alone or in combination with calcium-free phosphate binders might be a strategy to reduce phosphate levels and calcification and thus impact cardiovascular disease in CKD. METHODS We studied the effect of NA alone and in combination with magnesium carbonate (MgCO3) as a potential novel treatment strategy. CKD was induced in dilute brown non-agouti/2 mice by subtotal nephrectomy followed by a high-phosphate diet (HP) and 7 weeks of treatment with NA, MgCO3 or their combination. Control mice underwent subtotal nephrectomy and received an HP or underwent sham surgery and received standard chow plus NA. RESULTS CKD mice showed increased serum fibroblast growth factor 23 and calcium-phosphate product that was normalized by all treatment regimes. NA alone increased soft tissue and vascular calcification, whereas any treatment with MgCO3 significantly reduced calcification severity in CKD. While MgCO3 supplementation alone resulted in decreased calcification severity, it resulted in increased intestinal expression of the phosphate transporters type II sodium-dependent phosphate transporter 1 (Pit-1). Combined therapy of MgCO3 and NA reduced tissue calcification and normalized expression levels of intestinal phosphate transporter proteins. CONCLUSIONS In conclusion, the data indicate that NA increases while MgCO3 reduces ectopic calcification severity. Augmented expression of intestinal phosphate transporters by MgCO3 treatment was abolished by the addition of NA. However, the clinical relevance of the latter remains to be explored. Importantly, the data suggest no benefit of NA regarding treatment of calcification in addition to MgCO3.
Collapse
Affiliation(s)
- Nadine Kaesler
- Department of Nephrology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Claudia Goettsch
- Department of Cardiology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Daniel Weis
- Department of Nephrology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Leon Schurgers
- Department of Nephrology, University Hospital of the RWTH Aachen, Aachen, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
| | | | - Jürgen Floege
- Department of Nephrology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Rafael Kramann
- Department of Nephrology, University Hospital of the RWTH Aachen, Aachen, Germany.,Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW An overwhelming majority of chemotherapy agents are known to cause gastrointestinal mucositis, an unwanted side effect of cancer treatment, for which no effective treatment currently exists. The pathological processes underlying the development of gastrointestinal mucositis are many and varied, with multiple pathways thought to be involved in initiation of inflammation and apoptosis. Physiological and or biochemical-based deficiencies, such as vitamin D deficiency and gut microbiome density and population, are also thought to have an impact on mucositis severity. RECENT FINDINGS Recent studies investigating inflammatory pathways, such as cytokines and apoptotic markers, do show that interleukin-blocking proteins alleviate symptoms of gastrointestinal mucositis. However, the effectiveness of these treatments varies depending on the type of anticancer agent administered, meaning blocking compounds may be limited in their application. Targeting the host's gut microbiome in preventing dysbiosis is also thought to be a potential avenue for exploration. The use of probiotic gut bacteria (i.e. Lactobacillus spp.), while beneficial in preventing chemotherapy radiotherapy-induced diarrhoea, does not seem to alleviate the physiological damage caused by gastrointestinal mucositis. Vitamin D has been widely shown to have a host of anti-inflammatory and immunomodulatory effects in the intestine, as well as anticancer properties and therefore, may reduce severity of gastrointestinal mucositis. SUMMARY While anti-inflammatory and antiapoptotic agents have shown promise in animal models of gastrointestinal mucositis, there is still no singular mechanism allowing for the development of a therapeutic drug to prevent or cure gastrointestinal injury. A greater insight into the exact mechanistic actions of both probiotics and vitamin D might reveal how to improve their use as therapeutic treatments for gastrointestinal mucositis.
Collapse
|
34
|
Burns D, Werner A. Phosphate reabsorption in the kidney: NaPi-IIb or not IIb. Pflugers Arch 2020; 472:437-438. [PMID: 32236864 PMCID: PMC7165134 DOI: 10.1007/s00424-020-02374-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- David Burns
- Biosciences Institute, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Andreas Werner
- Biosciences Institute, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
35
|
Protective Role of Vitamin D in Renal Tubulopathies. Metabolites 2020; 10:metabo10030115. [PMID: 32204545 PMCID: PMC7142711 DOI: 10.3390/metabo10030115] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 02/06/2023] Open
Abstract
Vitamin D is tightly linked with renal tubular homeostasis: the mitochondria of proximal convoluted tubule cells are the production site of 1α,25-dihydroxyvitamin D3. Patients with renal impairment or tubular injury often suffer from chronic inflammation. This alteration comes from oxidative stress, acidosis, decreased clearance of inflammatory cytokines and stimulation of inflammatory factors. The challenge is to find the right formula for each patient to correctly modulate the landscape of treatment and preserve the essential functions of the organism without perturbating its homeostasis. The complexity of the counter-regulation mechanisms and the different axis involved in the Vitamin D equilibrium pose a major issue on Vitamin D as a potential effective anti-inflammatory drug. The therapeutic use of this compound should be able to inhibit the development of inflammation without interfering with normal homeostasis. Megalin-Cubilin-Amnionless and the FGF23-Klotho axis represent two Vitamin D-linked mechanisms that could modulate and ameliorate the damage response at the renal tubular level, balancing Vitamin D therapy with an effect potent enough to contrast the inflammatory cascades, but which avoids potential severe side effects.
Collapse
|
36
|
Cozzolino M, Ketteler M, Wagner CA. An expert update on novel therapeutic targets for hyperphosphatemia in chronic kidney disease: preclinical and clinical innovations. Expert Opin Ther Targets 2020; 24:477-488. [PMID: 32191548 DOI: 10.1080/14728222.2020.1743680] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: The management of hyperphosphatemia in patients with chronic kidney disease (CKD) is complicated, requiring a multidisciplinary approach that includes dietary phosphate restriction, dialysis, and phosphate binders.Areas covered: We describe key players involved in regulating inorganic phosphate homeostasis and their differential role in healthy people and different stages of CKD. The contribution of paracellular and transcellular intestinal absorptive mechanisms are also examined. Finally, we illuminate recent therapeutic approaches for hyperphosphatemia in CKD. We searched PubMed/Medline (up to November 2019) using the following terms: chronic kidney disease, dialysis, diet, hyperphosphatemia, NaPi2b, nicotinamide, phosphate binder, secondary hyperparathyroidism, tenapanor and vascular calcification.Expert opinion: The precise mechanisms regulating intestinal phosphate absorption in humans is not completely understood. However, it is now established that this process involves two independent pathways: a) active transport (i.e. transcellular route, via specific ion transporters) and inactive transport (i.e. paracellular route across tight junctions). Dietary phosphate restriction and phosphate-binder use can lead to an undesirable maladaptive increase in phosphate uptake and promote active phosphate transport by increased expression of the gastrointestinal sodium-dependent phosphate transporter, NaPi2b. Nicotinamide may overcome these limitations through the inhibition of NaPi2b, by improved efficacy and reduced phosphate binder use and better compliance.
Collapse
Affiliation(s)
- Mario Cozzolino
- Renal Division, ASST Santi Paolo E Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Markus Ketteler
- Department of General Internal Medicine and Nephrology, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Carsten Alexander Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,National Center of Competence in Research, NCCR Kidney. CH, Zurich, Switzerland
| |
Collapse
|
37
|
Omara II, Mou CT, Persia ME, Wong EA. Effects of available phosphorus source and concentration on performance and expression of sodium phosphate type IIb cotransporter, vitamin D-1α-hydroxylase, and vitamin D-24-hydroxylase mRNA in broiler chicks. Poult Sci 2020; 99:1822-1831. [PMID: 32241462 PMCID: PMC7587676 DOI: 10.1016/j.psj.2019.12.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/26/2019] [Accepted: 12/10/2019] [Indexed: 11/17/2022] Open
Abstract
This experiment was conducted to examine the effect of 2 phosphorus (P) sources on broiler performance to day 14. The P bioavailability was estimated using bird performance and tibia ash measurements, whereas P digestibility, intestinal P transporter, kidney vitamin D-1α-hydroxylase, and vitamin D-24-hydroxylase mRNA abundances were also determined. Slope regression analysis was used to determine the bioavailability of dicalcium phosphate (Dical P) and nanocalcium phosphate (Nano P) with dietary available P (AvP) set to 0.20% P (control) using AvP from the major ingredients and Dical P. The experimental treatments were achieved by supplementation with either Dical P or Nano P to generate 0.24, 0.28, 0.32, and 0.36% AvP. A total of 648-day-old unsexed broiler chicks were divided into 72 birds per treatment (8 replicate cages of 9 birds). Slope regression analysis showed positive linear relationships between BW, feed intake (FI), tibia ash weight (TAW), and tibia ash percentage (TAP) with dietary Dical P and Nano P levels. Comparisons between regression slopes for Dical P and Nano P fed birds were not significantly different for BW, feed intake, tibia ash weight, and tibia ash percentage, indicating similar P bioavailability from Dical P and Nano P. There were interactions between P source and AvP for feed efficiency (FE) and apparent ileal P digestibility (AIPD). Dicalcium phosphate had greater FE than Nano P at 0.28% AvP and greater AIPD than Nano P at 0.24% AvP. The addition of AvP from Dical P and Nano P resulted in reduced sodium phosphate cotransporter mRNA abundance in the duodenum in a dose–dependent response. In the kidney, vitamin D-1α-hydroxylase mRNA abundance was greater at 0.36% Nano P compared with control, but there was no difference with Dical P. There was no difference in vitamin D-24-hydroxylase mRNA abundance between control and supplementation with Nano P or Dical P. In conclusion, Nano P and Dical P had the same bioavailability but had different effects on gene expression.
Collapse
Affiliation(s)
- I I Omara
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24060, USA; Animal Production Department, Faculty of Agriculture, Cairo University, Giza 12613, Egypt
| | - C T Mou
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24060, USA
| | - M E Persia
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24060, USA
| | - E A Wong
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24060, USA.
| |
Collapse
|
38
|
Ryan BA, Kovacs CS. Calciotropic and phosphotropic hormones in fetal and neonatal bone development. Semin Fetal Neonatal Med 2020; 25:101062. [PMID: 31786156 DOI: 10.1016/j.siny.2019.101062] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
There are remarkable differences in bone and mineral metabolism between the fetus and adult. The fetal mineral supply is from active transport across the placenta. Calcium, phosphorus, and magnesium circulate at higher levels in the fetus compared to the mother. These high concentrations enable the skeleton to accrete required minerals before birth. Known key regulators in the adult include parathyroid hormone (PTH), calcitriol, fibroblast growth factor-23, calcitonin, and the sex steroids. But during fetal life, PTH plays a lesser role while the others appear to be unimportant. Instead, PTH-related protein (PTHrP) plays a critical role. After birth, serum calcium falls and phosphorus rises, which trigger an increase in PTH and a subsequent rise in calcitriol. The intestines become the main source of mineral supply while the kidneys reabsorb filtered minerals. This striking developmental switch is triggered by loss of the placenta, onset of breathing, and the drop in serum calcium.
Collapse
Affiliation(s)
- Brittany A Ryan
- Faculty of Medicine - Endocrinology, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Christopher S Kovacs
- Faculty of Medicine - Endocrinology, Memorial University of Newfoundland, St. John's, NL, Canada.
| |
Collapse
|
39
|
Majeed S, Qudsieh R, Edens FW, Brake J. Limestone particle size, calcium and phosphorus levels, and phytase effects on live performance and nutrients digestibility of broilers. Poult Sci 2020; 99:1502-1514. [PMID: 32111318 PMCID: PMC7587709 DOI: 10.1016/j.psj.2019.11.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/28/2019] [Accepted: 11/04/2019] [Indexed: 11/19/2022] Open
Abstract
Limestone particle size (PS) affects its solubility and thus can influence broiler performance by altering the rate of calcium (Ca) release into the gastrointestinal tract. The objective of this research was to determine, using 2 × 2 × 2 factorial arrangement, the influence of PS (fine and coarse) and supplemented phytase (0 and 1,000 FYT/kg) in diets formulated with 2 Ca and Pi levels (positive control [PC]; negative control [NC]) on live performance, bone ash, and apparent ileal nutrients digestibility (AID). Starter PC: 0.9 Ca and 0.45 Pi; NC: 0.72 Ca and 0.03 Pi. Grower PC: 0.76 Ca and 0.38 Pi; NC: 0.58 Ca and 0.23 Pi. The 8 diets were assigned randomly to a total of 1,512 birds, with 21 birds per pen and 9 pens per treatment. The main effects of PS and Ca and Pi levels had no influence on feed intake (FI), body weight gain (BWG), or feed conversion ratio. Adding phytase improved BWG by 8 g and 50 g and FI by 25 g and 56 g at 0–14 D (P ≤ 0.05) and 0–35 D (P ≤ 0.05), respectively. Interaction between Ca and Pi levels and phytase improved BWG and FI for 0–14 D (P ≤ 0.05) and BWG during 15–28 D (P ≤ 0.05) for PC without phytase and for PC and NC with phytase when compared with NC without phytase. Birds fed PC without phytase, or either PC or NC with phytase were about 96 g heavier than NC without phytase. Birds fed either PC or NC diet with coarse limestone or PC with fine limestone gained approximately 14 g more (P ≤ 0.05) than birds fed NC with fine limestone for BWG at 0–14 D (P ≤ 0.05). Phytase increased tibia bone ash (14 D) by 1% (P ≤ 0.05). AID of Ca and Pi at 14 D was improved (P ≤ 0.05) by 66% when phytase was added to coarse limestone. Results indicate that phytase improved broiler performance without being affected by PS. Furthermore, phytase had greater influence on coarse limestone than on fine limestone for bone ash and AID. Ca and Pi levels were the most influential factors in determining bone ash although phytase inclusion could lead to an improvement during early days.
Collapse
Affiliation(s)
- S Majeed
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695-7608
| | - R Qudsieh
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695-7608.
| | - F W Edens
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695-7608
| | - J Brake
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695-7608
| |
Collapse
|
40
|
Developmental Changes in Phosphate Homeostasis. Rev Physiol Biochem Pharmacol 2020; 179:117-138. [PMID: 33398502 DOI: 10.1007/112_2020_52] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Phosphate is a multivalent ion critical for a variety of physiological functions including bone formation, which occurs rapidly in the developing infant. In order to ensure maximal bone mineralization, young animals must maintain a positive phosphate balance. To accomplish this, intestinal absorption and renal phosphate reabsorption are greater in suckling and young animals relative to adults. This review discusses the known intestinal and renal adaptations that occur in young animals in order to achieve a positive phosphate balance. Additionally, we discuss the ontogenic changes in phosphotropic endocrine signalling as it pertains to intestinal and renal phosphate handling, including several endocrine factors not always considered in the traditional dogma of phosphotropic endocrine signalling, such as growth hormone, triiodothyronine, and glucocorticoids. Finally, a proposed model of how these factors may contribute to achieving a positive phosphate balance during development is proposed.
Collapse
|
41
|
Tay-Zar AC, Srichana P, Sadiq MB, Anal AK. Restriction of dietary non-phytate phosphorus on growth performance and expression of intestinal phosphate cotransporter genes in broilers. Poult Sci 2019; 98:4685-4693. [PMID: 30982064 DOI: 10.3382/ps/pez171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/13/2019] [Indexed: 11/20/2022] Open
Abstract
Effects of dietary non-phytate phosphorus (nPP) restriction on growth and duodenal type IIb sodium-dependent phosphate cotransporter (NaPi-IIb) genes were observed. A total of 432 one-day old Cobb500 male broiler chickens in 36 cage pens were divided into 6 groups with each group containing 6 pens. Each group was treated with one of the diets containing 0.33, 0.37, 0.41, 0.45, 0.49, and 0.53% of nPP up to 14 D. During 15 to 31 D, birds were treated with one of the diets containing 0.23, 0.27, 0.31, 0.35, 0.39, and 0.43% of nPP. Level of Ca was kept the same across all treatments. Dietary nPP level influenced (P < 0.001) weight gain and feed intake in both growth phases, whereas effect on feed per gain ratio was seen only in the second phase. Toe ash, tibia ash, and tibia breaking strength responded to treatments (P < 0.01) at 14 D. Only tibia ash content was significantly improved (P < 0.001) at 31 D. Growth and bone parameters linearly improved with an increase in dietary nPP content (P < 0.05). Above dietary nPP 0.41% and 0.31% for first phase and second phase, respectively, no significant improvement was seen. Duodenal NaPi-IIb mRNA overexpressed with a decrease in dietary nPP in both phases (P < 0.05). Relative expression of NaPi-IIb in lowest nPP group were 2.2 folds higher in the first phase and 3.6 folds higher in the second phase compared to respective highest nPP groups of each phase. No significant change in NaPi-IIb expression was seen above 0.37% of dietary nPP for 14 D and 0.31% of dietary nPP for 31 D. Dietary requirements of nPP 0.41% for 0 to 14 D and 0.31% for 15 to 31 D were adequate for optimal growth and bone parameters. This study fills the gap in understanding of intestinal NaPi-IIb expression in response to dietary nPP restriction in broilers older than 21 D of age.
Collapse
Affiliation(s)
- Aye Cho Tay-Zar
- Department of Food, Agriculture and BioResources, Asian Institute of Technology, Klong Luang 12120, Thailand
| | - Pairat Srichana
- Feed Technology Department, Charoen Pokphand Group, Bangrak, Bangkok 10500, Thailand
| | - Muhammad Bilal Sadiq
- Department of Food, Agriculture and BioResources, Asian Institute of Technology, Klong Luang 12120, Thailand
| | - Anil Kumar Anal
- Department of Food, Agriculture and BioResources, Asian Institute of Technology, Klong Luang 12120, Thailand
| |
Collapse
|
42
|
Korucu B, Helvaci O, Sadioglu R, Ozbas B, Yeter H, Derici U. Can Helicobacter pylori Colonization Affect the Phosphate Binder Pill Burden in Dialysis Patients? Ther Apher Dial 2019; 24:380-386. [PMID: 31622031 DOI: 10.1111/1744-9987.13443] [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: 08/11/2019] [Revised: 10/11/2019] [Accepted: 10/15/2019] [Indexed: 11/28/2022]
Abstract
Phosphate binder pill (PBP) burden is a significant problem in dialysis patients. Phosphate absorption through the paracellular pathway increases in relatively acidic pH. In this study, we evaluated the effect of factors contributing to duodenal pH-Helicobacter pylori (HP), proton pump inhibitors (PPIs), and NaHCO3 capsules-on PBP burden. We evaluated 255 dialysis patients with gastric biopsies and excluded patients with low Kt/V, gastrectomy, and parathyroidectomy. Patients were divided into groups and subgroups regarding HP existence, use of PPI, or NaHCO3 capsules. HP+ group had significantly higher PBP burden and PBP equivalent doses (P < 0.001; both). HP+ subgroup not using daily PPIs or NaHCO3 capsules had the highest PBP burden and PBP equivalent doses (P < 0.001; both). HP- subgroups had similar PBP and PBP equivalent doses (P = 0.446 and P = 0.382; respectively). HP colonization might affect the PBP burden in dialysis patients due to a decrease of duodenal pH.
Collapse
Affiliation(s)
- Berfu Korucu
- Department of Nephrology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Ozant Helvaci
- Department of Nephrology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Rezzan Sadioglu
- Department of Nephrology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Burak Ozbas
- Department of Nephrology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Hasan Yeter
- Department of Nephrology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Ulver Derici
- Department of Nephrology, Gazi University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
43
|
King AJ, Siegel M, He Y, Nie B, Wang J, Koo-McCoy S, Minassian NA, Jafri Q, Pan D, Kohler J, Kumaraswamy P, Kozuka K, Lewis JG, Dragoli D, Rosenbaum DP, O'Neill D, Plain A, Greasley PJ, Jönsson-Rylander AC, Karlsson D, Behrendt M, Strömstedt M, Ryden-Bergsten T, Knöpfel T, Pastor Arroyo EM, Hernando N, Marks J, Donowitz M, Wagner CA, Alexander RT, Caldwell JS. Inhibition of sodium/hydrogen exchanger 3 in the gastrointestinal tract by tenapanor reduces paracellular phosphate permeability. Sci Transl Med 2019; 10:10/456/eaam6474. [PMID: 30158152 DOI: 10.1126/scitranslmed.aam6474] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 10/31/2017] [Accepted: 03/06/2018] [Indexed: 12/16/2022]
Abstract
Hyperphosphatemia is common in patients with chronic kidney disease and is increasingly associated with poor clinical outcomes. Current management of hyperphosphatemia with dietary restriction and oral phosphate binders often proves inadequate. Tenapanor, a minimally absorbed, small-molecule inhibitor of the sodium/hydrogen exchanger isoform 3 (NHE3), acts locally in the gastrointestinal tract to inhibit sodium absorption. Because tenapanor also reduces intestinal phosphate absorption, it may have potential as a therapy for hyperphosphatemia. We investigated the mechanism by which tenapanor reduces gastrointestinal phosphate uptake, using in vivo studies in rodents and translational experiments on human small intestinal stem cell-derived enteroid monolayers to model ion transport physiology. We found that tenapanor produces its effect by modulating tight junctions, which increases transepithelial electrical resistance (TEER) and reduces permeability to phosphate, reducing paracellular phosphate absorption. NHE3-deficient monolayers mimicked the phosphate phenotype of tenapanor treatment, and tenapanor did not affect TEER or phosphate flux in the absence of NHE3. Tenapanor also prevents active transcellular phosphate absorption compensation by decreasing the expression of NaPi2b, the major active intestinal phosphate transporter. In healthy human volunteers, tenapanor (15 mg, given twice daily for 4 days) increased stool phosphorus and decreased urinary phosphorus excretion. We determined that tenapanor reduces intestinal phosphate absorption predominantly through reduction of passive paracellular phosphate flux, an effect mediated exclusively via on-target NHE3 inhibition.
Collapse
Affiliation(s)
| | | | - Ying He
- Ardelyx Inc., Fremont, CA 94555, USA
| | | | - Ji Wang
- Ardelyx Inc., Fremont, CA 94555, USA
| | | | | | | | - Deng Pan
- Ardelyx Inc., Fremont, CA 94555, USA
| | | | | | | | | | | | | | - Debbie O'Neill
- University of Alberta, Edmonton, Alberta T6G 1C9, Canada
| | - Allein Plain
- University of Alberta, Edmonton, Alberta T6G 1C9, Canada
| | - Peter J Greasley
- Cardiovascular and Metabolic Disease (CVMD) Translational Medicine Unit, Early Clinical Development, Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca Gothenburg, 431 50 Mölndal, Sweden
| | | | - Daniel Karlsson
- Bioscience, CVMD, IMED Biotech Unit, AstraZeneca Gothenburg, 431 50 Mölndal, Sweden
| | - Margareta Behrendt
- Bioscience, CVMD, IMED Biotech Unit, AstraZeneca Gothenburg, 431 50 Mölndal, Sweden
| | - Maria Strömstedt
- Bioscience, CVMD, IMED Biotech Unit, AstraZeneca Gothenburg, 431 50 Mölndal, Sweden
| | | | - Thomas Knöpfel
- Institute of Physiology, University of Zurich and National Center of Competence in Research Kidney Control of Homeostasis, CH-8057 Zurich, Switzerland
| | - Eva M Pastor Arroyo
- Institute of Physiology, University of Zurich and National Center of Competence in Research Kidney Control of Homeostasis, CH-8057 Zurich, Switzerland
| | - Nati Hernando
- Institute of Physiology, University of Zurich and National Center of Competence in Research Kidney Control of Homeostasis, CH-8057 Zurich, Switzerland
| | - Joanne Marks
- Department of Neuroscience, Physiology and Pharmacology, University College London, Royal Free Campus, London NW3 2PF, UK
| | - Mark Donowitz
- Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich and National Center of Competence in Research Kidney Control of Homeostasis, CH-8057 Zurich, Switzerland
| | | | | |
Collapse
|
44
|
Younes M, Aquilina G, Castle L, Engel KH, Fowler P, Frutos Fernandez MJ, Fürst P, Gürtler R, Husøy T, Mennes W, Moldeus P, Oskarsson A, Shah R, Waalkens-Berendsen I, Wölfle D, Aggett P, Cupisti A, Fortes C, Kuhnle G, Lillegaard IT, Scotter M, Giarola A, Rincon A, Tard A, Gundert-Remy U. Re-evaluation of phosphoric acid-phosphates - di-, tri- and polyphosphates (E 338-341, E 343, E 450-452) as food additives and the safety of proposed extension of use. EFSA J 2019; 17:e05674. [PMID: 32626329 PMCID: PMC7009158 DOI: 10.2903/j.efsa.2019.5674] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The Panel on Food Additives and Flavourings added to Food (FAF) provided a scientific opinion re-evaluating the safety of phosphates (E 338-341, E 343, E 450-452) as food additives. The Panel considered that adequate exposure and toxicity data were available. Phosphates are authorised food additives in the EU in accordance with Annex II and III to Regulation (EC) No 1333/2008. Exposure to phosphates from the whole diet was estimated using mainly analytical data. The values ranged from 251 mg P/person per day in infants to 1,625 mg P/person per day for adults, and the high exposure (95th percentile) from 331 mg P/person per day in infants to 2,728 mg P/person per day for adults. Phosphate is essential for all living organisms, is absorbed at 80-90% as free orthophosphate excreted via the kidney. The Panel considered phosphates to be of low acute oral toxicity and there is no concern with respect to genotoxicity and carcinogenicity. No effects were reported in developmental toxicity studies. The Panel derived a group acceptable daily intake (ADI) for phosphates expressed as phosphorus of 40 mg/kg body weight (bw) per day and concluded that this ADI is protective for the human population. The Panel noted that in the estimated exposure scenario based on analytical data exposure estimates exceeded the proposed ADI for infants, toddlers and other children at the mean level, and for infants, toddlers, children and adolescents at the 95th percentile. The Panel also noted that phosphates exposure by food supplements exceeds the proposed ADI. The Panel concluded that the available data did not give rise to safety concerns in infants below 16 weeks of age consuming formula and food for medical purposes.
Collapse
|
45
|
Saurette M, Alexander RT. Intestinal phosphate absorption: The paracellular pathway predominates? Exp Biol Med (Maywood) 2019; 244:646-654. [PMID: 30764666 DOI: 10.1177/1535370219831220] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
IMPACT STATEMENT This review summarizes the work on transcellular intestinal phosphate absorption, arguing why this pathway is not the predominant pathway in humans consuming a "Western" diet. We then highlight the recent evidence which is strongly consistent with paracellular intestinal phosphate absorption mediating the bulk of intestinal phosphate absorption in humans.
Collapse
Affiliation(s)
- Matthew Saurette
- 1 Department of Physiology, University of Alberta, Edmonton, Alberta T6G 2R7, Canada.,2 The Women's & Children's Health Research Institute, Edmonton, Alberta T6G 1C9, Canada
| | - R Todd Alexander
- 1 Department of Physiology, University of Alberta, Edmonton, Alberta T6G 2R7, Canada.,2 The Women's & Children's Health Research Institute, Edmonton, Alberta T6G 1C9, Canada.,3 Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2R7, Canada
| |
Collapse
|
46
|
Abstract
Calcium kidney stones are common worldwide. Most are idiopathic and composed of calcium oxalate. Calcium phosphate is present in around 80% and may initiate stone formation. Stone production is multifactorial with a polygenic genetic contribution. Phosphaturia is found frequently among stone formers but until recently received scant attention. This review examines possible mechanisms for the phosphaturia and its relevance to stone formation from a wide angle. There is a striking lack of clinical data. Phosphaturia is associated, but not correlated, with hypercalciuria, increased 1,25 dihydroxy-vitamin D [1,25 (OH)2D], and sometimes evidence of disturbances in proximal renal tubular function. Phosphate reabsorption in the proximal renal tubules requires tightly regulated interaction of many proteins. Paracellular flow through intercellular tight junctions is the major route of phosphate absorption from the intestine and can be reduced therapeutically in hyperphosphatemic patients. In monogenic defects stones develop when phosphaturia is associated with hypercalciuria, generally explained by increased 1,25 (OH)2D production in response to hypophosphatemia. Calcification does not occur in disorders with increased FGF23 when phosphaturia occurs in isolation and 1,25 (OH)2D is suppressed. Candidate gene studies have identified mutations in the phosphate transporters, but in few individuals. One genome-wide study identified a polymorphism of the phosphate transporter gene SLC34A4 associated with stones. Others did not find mutations obviously linked to phosphate reabsorption. Future genetic studies should have a wide trawl and should focus initially on groups of patients with clearly defined phenotypes. The global data should be pooled.
Collapse
Affiliation(s)
- Valerie Walker
- Department of Clinical Biochemistry, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.
| |
Collapse
|
47
|
Yin Z, Du J, Yu F, Xia W. Tumor-induced osteomalacia. Osteoporos Sarcopenia 2018; 4:119-127. [PMID: 30775554 PMCID: PMC6372818 DOI: 10.1016/j.afos.2018.12.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/02/2018] [Accepted: 12/04/2018] [Indexed: 12/30/2022] Open
Abstract
Tumor-induced osteomalacia (TIO), also known as oncogenic osteomalacia, is a rare paraneoplastic syndrome characterized by hypophosphatemia resulting from decreased tubular phosphate reabsorption, with a low or inappropriately normal level of active vitamin D. The culprit tumors of TIO could produce fibroblast growth factor 23 which plays a role in regulating renal Pi handling and 25-hydroxyvitamin D 1α-hydroxylase activity. Chronic hypophosphatemia could eventually lead to inadequate bone mineralization, presenting as osteomalacia. The diagnosis should be considered when patients manifest as hypophosphatemia and osteomalacia, or rickets and needs to be differentiated from other disorders of phosphate metabolism, such as the inhereditary diseases like X-linked hypophosphataemic rickets, autosomal dominant hypophosphataemic rickets, autosomal recessive hypophosphataemic rickets and acquired diseases like vitamin D deficiency. Localization of responsible tumors could be rather difficult since the vast majority are very small and could be everywhere in the body. A combination of thorough physical examination, laboratory tests and imaging techniques should be applied and sometimes a venous sampling may come into handy. The technology of somatostatin-receptor functional scintigraphy markedly facilitates the localization of TIO tumor. Patients undergoing complete removal of the causative neoplasm generally have favorable prognoses while a few have been reported to suffer from recurrence and metastasis. For those undetectable or unresectable cases, phosphate supplements and active vitamin D should be administrated and curative intended radiotherapy or ablation is optional.
Collapse
Affiliation(s)
| | | | | | - Weibo Xia
- Department of Endocrinology, Key Laboratory of Endocrinology, The National Commission of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
48
|
Hu MC, Shi M, Moe OW. Role of αKlotho and FGF23 in regulation of type II Na-dependent phosphate co-transporters. Pflugers Arch 2018; 471:99-108. [PMID: 30506274 DOI: 10.1007/s00424-018-2238-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/16/2018] [Accepted: 11/18/2018] [Indexed: 11/26/2022]
Abstract
Alpha-Klotho is a member of the Klotho family consisting of two other single-pass transmembrane proteins: βKlotho and γKlotho; αKlotho has been shown to circulate in the blood. Fibroblast growth factor (FGF)23 is a member of the FGF superfamily of 22 genes/proteins. αKlotho serves as a co-receptor with FGF receptors (FGFRs) to provide a receptacle for physiological FGF23 signaling including regulation of phosphate metabolism. The extracellular domain of transmembrane αKlotho is shed by secretases and released into blood circulation (soluble αKlotho). Soluble αKlotho has both FGF23-independent and FGF23-dependent roles in phosphate homeostasis by modulating intestinal phosphate absorption, urinary phosphate excretion, and phosphate distribution into bone in concerted interaction with other calciophosphotropic hormones such as PTH and 1,25-(OH)2D. The direct role of αKlotho and FGF23 in the maintenance of phosphate homeostasis is partly mediated by modulation of type II Na+-dependent phosphate co-transporters in target organs. αKlotho and FGF23 are principal phosphotropic hormones, and the manipulation of the αKlotho-FGF23 axis is a novel therapeutic strategy for genetic and acquired phosphate disorders and for conditions with FGF23 excess and αKlotho deficiency such as chronic kidney disease.
Collapse
Affiliation(s)
- Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Mingjun Shi
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
49
|
Vorland CJ, Lachcik PJ, Aromeh LO, Moe SM, Chen NX, Hill Gallant KM. Effect of dietary phosphorus intake and age on intestinal phosphorus absorption efficiency and phosphorus balance in male rats. PLoS One 2018; 13:e0207601. [PMID: 30452474 PMCID: PMC6242370 DOI: 10.1371/journal.pone.0207601] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 10/26/2018] [Indexed: 12/31/2022] Open
Abstract
Intestinal phosphorus absorption is an important component of whole-body phosphorus metabolism, and limiting dietary phosphorus absorption is particularly of interest as a therapeutic target in patients with chronic kidney disease to manage mineral bone disorders. Yet, mechanisms and regulation of intestinal phosphorus absorption have not been adequately studied and discrepancies in findings exist based on the absorption assessment technique used. In vitro techniques show rather consistent effects of dietary phosphorus intake level and age on intestinal sodium-dependent phosphate transport. But, the few studies that have used in vivo techniques conflict with these in vitro studies. Therefore, we aimed to investigate the effects of dietary phosphorus intake level on phosphorus absorption using the in situ ligated loop technique in three different aged rats. Male Sprague-Dawley rats (n = 72), were studied at 10-, 20-, and 30-weeks-of-age on a low (0.1%), normal (0.6%), or high (1.2%) phosphorus diet in a 3x3 factorial design (n = 8/group). Rats were fed their assigned diet for 2-weeks prior to absorption testing by jejunal ligated loop as a non-survival procedure, utilizing 33P radioisotope. Metabolic cages were used for determination of calcium and phosphorus balance over the final four days prior to sacrifice, and blood was collected at the time of sacrifice for biochemistries. Our results show that phosphorus absorption was higher in 10-week-old rats compared with 20- and 30-week-olds and this corresponded to higher gene expression of the major phosphate transporter, NaPi-2b, as well as higher whole-body phosphorus balance and net phosphorus absorption. Dietary phosphorus intake level did not affect jejunal phosphorus absorption or NaPi-2b gene expression. Our results contrast with studies utilizing in vitro techniques, but corroborate results of other rodent studies utilizing in situ or in vivo methods. Thus, there is need for additional studies that employ more physiological methods of phosphorus absorption assessment.
Collapse
Affiliation(s)
- Colby J. Vorland
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States of America
| | - Pamela J. Lachcik
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States of America
| | - Loretta O. Aromeh
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Sharon M. Moe
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States of America
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States of America
- Department of Medicine, Roudebush Veterans Affairs Medicine Center, Indianapolis, IN, United States of America
| | - Neal X. Chen
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Kathleen M. Hill Gallant
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States of America
- * E-mail:
| |
Collapse
|
50
|
Thomas L, Xue J, Dominguez Rieg JA, Rieg T. Contribution of NHE3 and dietary phosphate to lithium pharmacokinetics. Eur J Pharm Sci 2018; 128:1-7. [PMID: 30419292 DOI: 10.1016/j.ejps.2018.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/09/2018] [Accepted: 11/08/2018] [Indexed: 01/13/2023]
Abstract
Lithium is one of the mainstays for the treatment of bipolar disorder despite its side effects on the endocrine, neurological, and renal systems. Experimentally, lithium has been used as a measure to determine proximal tubule reabsorption based on the assumption that lithium and sodium transport go in parallel in the proximal tubule. However, the exact mechanism by which lithium is reabsorbed remains elusive. The majority of proximal tubule sodium reabsorption is directly or indirectly mediated by the sodium-hydrogen exchanger 3 (NHE3). In addition, sodium-phosphate cotransporters have been implicated in renal lithium reabsorption. In order to better understand the role of sodium-phosphate cotransporters involved in lithium (re)absorption, we studied lithium pharmacokinetics in: i) tubule-specific NHE3 knockout mice (NHE3loxloxPax8Cre), and ii) mice challenged with low or high phosphate diets. Intravenous or oral administration of lithium did not result in differences in lithium bioavailability, half-life, maximum plasma concentrations, area under the curve, lithium clearance, or urinary lithium/creatinine ratios between control and NHE3loxloxPax8Cre mice. After one week of dietary phosphate challenges, lithium bioavailability was ~30% lower on low versus high dietary phosphate, possibly the consequence of a smaller area under the curve after oral administration. This was associated with higher apparent lithium clearance after oral administration and lower urinary lithium/creatinine ratios on low versus high dietary phosphate. Collectively, renal NHE3 does not play a role in lithium pharmacokinetics; however, dietary phosphate could have an indirect effect on lithium bioavailability and lithium disposition.
Collapse
Affiliation(s)
- Linto Thomas
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33612, USA
| | - Jianxiang Xue
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33612, USA
| | - Jessica A Dominguez Rieg
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33612, USA
| | - Timo Rieg
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|