51
|
Johansson S, Rosenbaum DP, Palm J, Stefansson B, Knutsson M, Lisbon EA, Hilgendorf C. Tenapanor administration and the activity of the H + -coupled transporter PepT1 in healthy volunteers. Br J Clin Pharmacol 2017; 83:2008-2014. [PMID: 28432691 PMCID: PMC5582369 DOI: 10.1111/bcp.13313] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 04/06/2017] [Accepted: 04/18/2017] [Indexed: 12/24/2022] Open
Abstract
AIM Tenapanor (RDX5791/AZD1722), an inhibitor of gastrointestinal Na+ /H+ exchanger NHE3, is being evaluated for the treatment of patients with constipation-predominant irritable bowel syndrome and the treatment of hyperphosphataemia in patients with chronic kidney disease on dialysis. By reducing intestinal H+ secretion, inhibition of NHE3 by tenapanor could indirectly affect H+ -coupled transporter activity, leading to drug-drug interactions. We investigated the effect of tenapanor on the activity of the H+ -coupled peptide transporter PepT1 via assessment of the pharmacokinetics of cefadroxil - a compound transported by PepT1 - in healthy volunteers. METHODS In this open-label, two-period crossover, phase 1 study (NCT02140281), 28 volunteers received in random order: a single dose of cefadroxil 500 mg for 1 day; and tenapanor 15 mg twice daily over 4 days followed by single doses of both cefadroxil 500 mg and tenapanor 15 mg on day 5. There was a 4-day washout between treatment periods. RESULTS Cefadroxil exposure was similar when administered alone or in combination with tenapanor {geometric least-squares mean ratios [(cefadroxil + tenapanor)/cefadroxil] (90% confidence interval): area under the concentration-time curve 93.3 (90.6-96.0)%; maximum concentration in plasma 95.9 (89.8-103)%}. Tenapanor treatment caused a softening of stool consistency and an increase in stool frequency, consistent with its expected pharmacodynamic effect. No safety concerns were identified and tenapanor was not detected in plasma. CONCLUSIONS These results suggest that tenapanor 15 mg twice daily does not have a clinically relevant impact on the activity of the H+ -coupled transporter PepT1 in humans. This may guide future research on drug-drug interactions involving NHE3 inhibitors.
Collapse
Affiliation(s)
- Susanne Johansson
- Quantitative Clinical Pharmacology, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden
| | | | - Johan Palm
- Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Mölndal, Sweden
| | - Bergur Stefansson
- Cardiovascular Metabolic Diseases, Global Medicines Development, AstraZeneca, Mölndal, Sweden
| | - Mikael Knutsson
- Biometrics & Information Sciences, Global Medicines Development, AstraZeneca, Mölndal, Sweden
| | | | - Constanze Hilgendorf
- Drug Metabolism and Pharmacokinetics, Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden
| |
Collapse
|
52
|
Beggs MR, Alexander RT. Intestinal absorption and renal reabsorption of calcium throughout postnatal development. Exp Biol Med (Maywood) 2017; 242:840-849. [PMID: 28346014 DOI: 10.1177/1535370217699536] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Calcium is vital for many physiological functions including bone mineralization. Postnatal deposition of calcium into bone is greatest in infancy and continues through childhood and adolescence until peek mineral density is reached in early adulthood. Thereafter, bone mineral density remains static until it eventually declines in later life. A positive calcium balance, i.e. more calcium absorbed than excreted, is crucial to bone deposition during growth and thus to peek bone mineral density. Dietary calcium is absorbed from the intestine into the blood. It is then filtered by the renal glomerulus and either reabsorbed by the tubule or excreted in the urine. Calcium can be (re)absorbed across intestinal and renal epithelia via both transcellular and paracellular pathways. Current evidence suggests that significant intestinal and renal calcium transport changes occur throughout development. However, the molecular details of these alterations are incompletely delineated. Here we first briefly review the current model of calcium transport in the intestine and renal tubule in the adult. Then, we describe what is known with regard to calcium handling through postnatal development, and how alterations may aid in mediating a positive calcium balance. The role of transcellular and paracellular calcium transport pathways and the contribution of specific intestinal and tubular segments vary with age. However, the current literature highlights knowledge gaps in how specifically intestinal and renal calcium (re)absorption occurs early in postnatal development. Future research should clarify the specific changes in calcium transport throughout early postnatal development including mediators of these alterations enabling appropriate bone mineralization. Impact statement This mini review outlines the current state of knowledge pertaining to the molecules and mechanisms maintaining a positive calcium balance throughout postnatal development. This process is essential to achieving optimal bone mineral density in early adulthood, thereby lowering the lifetime risk of osteoporosis.
Collapse
Affiliation(s)
- Megan R Beggs
- 1 Department of Physiology, University of Alberta, Edmonton, Alberta T6G 2R7, Canada
| | - R Todd Alexander
- 1 Department of Physiology, University of Alberta, Edmonton, Alberta T6G 2R7, Canada.,2 Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2R7, Canada
| |
Collapse
|
53
|
Wang YY, Lin YH, Wu YN, Chen YL, Lin YC, Cheng CY, Chiang HS. Loss of SLC9A3 decreases CFTR protein and causes obstructed azoospermia in mice. PLoS Genet 2017; 13:e1006715. [PMID: 28384194 PMCID: PMC5398719 DOI: 10.1371/journal.pgen.1006715] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 04/20/2017] [Accepted: 03/24/2017] [Indexed: 01/21/2023] Open
Abstract
Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause cystic fibrosis (CF) and are associated with congenital bilateral absence of the vas deferens (CBAVD), which is the major cause of infertility in male patients with CF. However, most Taiwanese patients with CBAVD do not carry major CFTR mutations. Some patients have a single copy deletion of the solute carrier family 9 isoform 3 (SLC9A3) gene. SLC9A3 is a Na+/H+ exchanger, and depleted Slc9a3 in male mice causes infertility due to the abnormal dilated lumen of the rete testis and efferent ductules. Furthermore, SLC9A3 interacts with CFTR in the pancreatic duct and functions as a genetic modifier of CF. However, SLC9A3 function and its relation to CFTR expression in the male reproductive tract in vivo remain elusive. In the present study, we found that CFTR expression was dramatically decreased in the epididymis and vas deferens of Slc9a3 knockout mice. Adult Slc9a3-/- mice showed not only significantly decreased epididymis and vas deferens weight but also increased testis weight. Furthermore, Slc9a3-/- mice developed obstructive azoospermia because of abnormal abundant secretions and calcification in the lumen of the reproductive tract. Ultrastructural analysis of the epithelium in Slc9a3–/–epididymis and vas deferens displayed disorganized and reduced number of stereocilia and numerous secretory apparatuses. Our data revealed that interdependence between SLC9A3 and CFTR is critical for maintaining a precise microenvironment in the epithelial cytoarchitecture of the male reproductive tract. The Slc9a3-deficient mice with impaired male excurrent ducts in this study provide proof for our clinical findings that some Taiwanese of CBAVD carry SLC9A3 deletion but without major CFTR mutations. Cystic fibrosis (CF) is the most common inherited life-threatening disease in Caucasians. The most well-known cause of CF is a genetic defect in CFTR, an apical membrane chloride and bicarbonate channel. The symptoms of CF include defects in the respiratory, digestive, and male reproductive systems. Most male patients with CF are infertile due to congenital bilateral absence of the vas deferens (CBAVD), which leads to obstructive azoospermia. Nevertheless, Taiwanese patients with CBAVD do not carry the common mutations of CFTR found in Caucasians. We have identified a potential candidate, SLC9A3, of which a single copy is lost in Taiwanese patients with CBAVD. In addition to the previously reported role of SLC9A3 in the digestive system and efferent ductules, we now report that the SLC9A3 deficiency causes obstructive azoospermia and impairs the epithelial structure of the reproductive tract. Loss of SLC9A3 also leads to dramatic reduced expression of CFTR in the reproductive tract. We suggest that the interplay between SLC9A3 and CFTR is responsible for CF-related infertility. Thus, we have characterized a potential critical player in the pathogenesis of CBAVD and provide a new diagnostic candidate for Asian patients with CBAVD.
Collapse
Affiliation(s)
- Ya-Yun Wang
- Department of Chemistry, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Ying-Hung Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Yi-No Wu
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Yen-Lin Chen
- Department of Chemistry, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
- Department of Pathology, Cardinal Tien Hospital, New Taipei City, Taiwan
| | - Yung-Chih Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chiao-Yin Cheng
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Han-Sun Chiang
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, Taiwan
- Department of Urology, Taipei Medical University Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
54
|
Fenton RA, Poulsen SB, de la Mora Chavez S, Soleimani M, Dominguez Rieg JA, Rieg T. Renal tubular NHE3 is required in the maintenance of water and sodium chloride homeostasis. Kidney Int 2017; 92:397-414. [PMID: 28385297 DOI: 10.1016/j.kint.2017.02.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 01/25/2017] [Accepted: 02/02/2017] [Indexed: 11/30/2022]
Abstract
The sodium/proton exchanger isoform 3 (NHE3) is expressed in the intestine and the kidney, where it facilitates sodium (re)absorption and proton secretion. The importance of NHE3 in the kidney for sodium chloride homeostasis, relative to the intestine, is unknown. Constitutive tubule-specific NHE3 knockout mice (NHE3loxloxCre) did not show significant differences compared to control mice in body weight, blood pH or bicarbonate and plasma sodium, potassium, or aldosterone levels. Fluid intake, urinary flow rate, urinary sodium/creatinine, and pH were significantly elevated in NHE3loxloxCre mice, while urine osmolality and GFR were significantly lower. Water deprivation revealed a small urinary concentrating defect in NHE3loxloxCre mice on a control diet, exaggerated on low sodium chloride. Ten days of low or high sodium chloride diet did not affect plasma sodium in control mice; however, NHE3loxloxCre mice were susceptible to low sodium chloride (about -4 mM) or high sodium chloride intake (about +2 mM) versus baseline, effects without differences in plasma aldosterone between groups. Blood pressure was significantly lower in NHE3loxloxCre mice and was sodium chloride sensitive. In control mice, the expression of the sodium/phosphate co-transporter Npt2c was sodium chloride sensitive. However, lack of tubular NHE3 blunted Npt2c expression. Alterations in the abundances of sodium/chloride cotransporter and its phosphorylation at threonine 58 as well as the abundances of the α-subunit of the epithelial sodium channel, and its cleaved form, were also apparent in NHE3loxloxCre mice. Thus, renal NHE3 is required to maintain blood pressure and steady-state plasma sodium levels when dietary sodium chloride intake is modified.
Collapse
Affiliation(s)
- Robert A Fenton
- InterPrET Center, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Søren B Poulsen
- InterPrET Center, Department of Biomedicine, Aarhus University, Aarhus, Denmark; VA San Diego Healthcare System, San Diego, California, USA
| | | | - Manoocher Soleimani
- Department of Medicine, University of Cincinnati, Cincinnati, Ohio, USA; Research Services, Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Jessica A Dominguez Rieg
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA
| | - Timo Rieg
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA.
| |
Collapse
|
55
|
Thammayon N, Wongdee K, Lertsuwan K, Suntornsaratoon P, Thongbunchoo J, Krishnamra N, Charoenphandhu N. Na +/H + exchanger 3 inhibitor diminishes the amino-acid-enhanced transepithelial calcium transport across the rat duodenum. Amino Acids 2017; 49:725-734. [PMID: 27981415 DOI: 10.1007/s00726-016-2374-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 11/29/2016] [Indexed: 12/22/2022]
Abstract
Na+/H+ exchanger (NHE)-3 is important for intestinal absorption of nutrients and minerals, including calcium. The previous investigations have shown that the intestinal calcium absorption is also dependent on luminal nutrients, but whether aliphatic amino acids and glucose, which are abundant in the luminal fluid during a meal, similarly enhance calcium transport remains elusive. Herein, we used the in vitro Ussing chamber technique to determine epithelial electrical parameters, i.e., potential difference (PD), short-circuit current (Isc), and transepithelial resistance, as well as 45Ca flux in the rat duodenum directly exposed on the mucosal side to glucose or various amino acids. We found that mucosal glucose exposure led to the enhanced calcium transport, PD, and Isc, all of which were insensitive to NHE3 inhibitor (100 nM tenapanor). In the absence of mucosal glucose, several amino acids (12 mM in the mucosal side), i.e., alanine, isoleucine, leucine, proline, and hydroxyproline, markedly increased the duodenal calcium transport. An inhibitor for NHE3 exposure on the mucosal side completely abolished proline- and leucine-enhanced calcium transport, but not transepithelial transport of both amino acids themselves. In conclusion, glucose and certain amino acids in the mucosal side were potent stimulators of the duodenal calcium absorption, but only amino-acid-enhanced calcium transport was NHE3-dependent.
Collapse
Affiliation(s)
- Nithipak Thammayon
- Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Kannikar Wongdee
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Faculty of Allied Health Sciences, Burapha University, Chonburi, 20131, Thailand
| | - Kornkamon Lertsuwan
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Department of Biochemistry, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
| | - Panan Suntornsaratoon
- Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Jirawan Thongbunchoo
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Nateetip Krishnamra
- Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Narattaphol Charoenphandhu
- Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand.
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, 73170, Thailand.
| |
Collapse
|
56
|
Ure ME, Heydari E, Pan W, Ramesh A, Rehman S, Morgan C, Pinsk M, Erickson R, Herrmann JM, Dimke H, Cordat E, Lemaire M, Walter M, Alexander RT. A variant in a cis-regulatory element enhances claudin-14 expression and is associated with pediatric-onset hypercalciuria and kidney stones. Hum Mutat 2017; 38:649-657. [PMID: 28229505 DOI: 10.1002/humu.23202] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 01/26/2017] [Accepted: 02/18/2017] [Indexed: 12/19/2022]
Abstract
The greatest risk factor for kidney stones is hypercalciuria, the etiology of which is largely unknown. A recent genome-wide association study (GWAS) linked hypercalciuria and kidney stones to a claudin-14 (CLDN14) risk haplotype. However, the underlying molecular mechanism was not delineated. Recently, renal CLDN14 expression was found to increase in response to increased plasma calcium, thereby inducing calciuria. We hypothesized therefore that some children with hypercalciuria and kidney stones harbor a CLDN14 variant that inappropriately increases gene expression. To test this hypothesis, we sequenced the CLDN14 risk haplotype in a cohort of children with idiopathic hypercalciuria and kidney stones. An intronic SNP was more frequent in affected children. Dual luciferase and cell-based assays demonstrated increased reporter or CLDN14 expression when this polymorphism was introduced. In silico studies predicted the SNP introduced a novel insulinoma-associated 1 (INSM1) transcription factor binding site. Consistent with this, repeating the dual luciferase assay in the presence of INSM1 further increased reporter expression. Our data suggest that children with the INSM1 binding site within the CLDN14 risk haplotype have a higher likelihood of hypercalciuria and kidney stones. Enhanced CLDN14 expression may play a role in the pathophysiology of their hypercalciuria.
Collapse
Affiliation(s)
- Megan E Ure
- Department of Pediatrics, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| | - Emma Heydari
- Department of Pediatrics, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| | - Wanling Pan
- Department of Physiology, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| | - Ajay Ramesh
- Depratment of Cell Biology, University of Kaiserslautern, Kaiserslautern, 67663, Germany
| | - Sabah Rehman
- Department of Pediatrics, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| | - Catherine Morgan
- Department of Pediatrics, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| | - Maury Pinsk
- Department of Pediatrics, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| | - Robin Erickson
- Department of Pediatrics, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 0W8, Canada
| | - Johannes M Herrmann
- Depratment of Cell Biology, University of Kaiserslautern, Kaiserslautern, 67663, Germany
| | - Henrik Dimke
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, 5000, Denmark
| | - Emmanuelle Cordat
- Department of Pediatrics, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| | - Mathieu Lemaire
- The Hospital for Sick Children, Toronto, Ontario, M5G 1×8, Canada
| | - Michael Walter
- Department of Medical Genetics, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| | - R Todd Alexander
- Department of Pediatrics, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada.,Department of Physiology, The University of Alberta, Edmonton, Alberta, T6G 2R7, Canada
| |
Collapse
|
57
|
Charoenphandhu N, Kraidith K, Lertsuwan K, Sripong C, Suntornsaratoon P, Svasti S, Krishnamra N, Wongdee K. Na +/H + exchanger 3 inhibitor diminishes hepcidin-enhanced duodenal calcium transport in hemizygous β-globin knockout thalassemic mice. Mol Cell Biochem 2016; 427:201-208. [PMID: 27995414 DOI: 10.1007/s11010-016-2911-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/03/2016] [Indexed: 12/15/2022]
Abstract
Recent investigation has shown that the liver-derived iron-regulating hormone, hepcidin, can potentiate intestinal calcium absorption in hemizygous β-globin knockout thalassemic (BKO) mice. Since the upregulation of Fe2+ and H+ cotransporter, divalent metal transporter (DMT)-1, has been shown to correlate with thalassemia-induced intestinal calcium absorption impairment, the inhibition of the apical Na+/H+ exchanger (NHE)-3 that is essential for cytoplasmic pH regulation and transepithelial sodium absorption was hypothesized to negatively affect hepcidin action. Herein, the positive effect of hepcidin on the duodenal calcium transport was evaluated using Ussing chamber technique. The results showed that BKO mice had lower absorptive surface area and duodenal calcium transport than wild-type mice. Besides, paracellular transport of zinc in BKO mice was compromised. Hepcidin administration completely restored calcium transport. Since this hepcidin action was totally abolished by inhibitors of the basolateral calcium transporters, Na+/Ca2+ exchanger (NCX1) and plasma membrane Ca2+-ATPase (PMCA1b), the enhanced calcium flux potentially occurred through the transcellular pathway rather than paracellular pathway. Interestingly, the selective NHE3 inhibitor, 100 nM tenapanor, markedly inhibited hepcidin-enhanced calcium transport. Accordingly, hepcidin is one of the promising therapeutic agents for calcium malabsorption in β-thalassemia. It mainly stimulates the transcellular calcium transport across the duodenal epithelium in an NHE3-dependent manner.
Collapse
Affiliation(s)
- Narattaphol Charoenphandhu
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand.,Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand.,Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Kamonshanok Kraidith
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand.,Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Kornkamon Lertsuwan
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand.,Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Chanakarn Sripong
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand.,Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Panan Suntornsaratoon
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand.,Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Saovaros Svasti
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Nateetip Krishnamra
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand.,Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Kannikar Wongdee
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand. .,Office of Academic Management, Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand.
| |
Collapse
|
58
|
Alexander RT, Cordat E, Chambrey R, Dimke H, Eladari D. Acidosis and Urinary Calcium Excretion: Insights from Genetic Disorders. J Am Soc Nephrol 2016; 27:3511-3520. [PMID: 27468975 DOI: 10.1681/asn.2016030305] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Metabolic acidosis is associated with increased urinary calcium excretion and related sequelae, including nephrocalcinosis and nephrolithiasis. The increased urinary calcium excretion induced by metabolic acidosis predominantly results from increased mobilization of calcium out of bone and inhibition of calcium transport processes within the renal tubule. The mechanisms whereby acid alters the integrity and stability of bone have been examined extensively in the published literature. Here, after briefly reviewing this literature, we consider the effects of acid on calcium transport in the renal tubule and then discuss why not all gene defects that cause renal tubular acidosis are associated with hypercalciuria and nephrocalcinosis.
Collapse
Affiliation(s)
- R Todd Alexander
- Departments of Pediatrics and .,Physiology, University of Alberta, Edmonton, Canada
| | | | - Régine Chambrey
- Institut National de la Santé et de la Recherche Médicale U970, Paris Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Henrik Dimke
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Demark; and
| | - Dominique Eladari
- Institut National de la Santé et de la Recherche Médicale U970, Paris Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Department of Physiologie, Hôpital Européen Georges Pompidou, Assistance Publique Hôpitaux de Paris, Paris, France
| |
Collapse
|
59
|
Granjon D, Bonny O, Edwards A. A model of calcium homeostasis in the rat. Am J Physiol Renal Physiol 2016; 311:F1047-F1062. [PMID: 27358053 DOI: 10.1152/ajprenal.00230.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 06/27/2016] [Indexed: 12/14/2022] Open
Abstract
We developed a model of calcium homeostasis in the rat to better understand the impact of dysfunctions such as primary hyperparathyroidism and vitamin D deficiency on calcium balance. The model accounts for the regulation of calcium intestinal uptake, bone resorption, and renal reabsorption by parathyroid hormone (PTH), vitamin D3, and Ca2+ itself. It is the first such model to incorporate recent findings regarding the role of the calcium-sensing receptor (CaSR) in the kidney, the presence of a rapidly exchangeable pool in bone, and the delayed response of vitamin D3 synthesis. Accounting for two (fast and slow) calcium storage compartments in bone allows the model to properly predict the effects of bisphophonates on the plasma levels of Ca2+ ([Ca2+]p), PTH, and vitamin D3 Our model also suggests that Ca2+ exchange rates between plasma and the fast pool vary with both sex and age, allowing [Ca2+]p to remain constant in spite of sex- and age-based hormonal and other differences. Our results suggest that the inconstant hypercalciuria that is observed in primary hyperparathyroidism can be attributed in part to counterbalancing effects of PTH and CaSR in the kidney. Our model also correctly predicts that calcimimetic agents such as cinacalcet bring down [Ca2+]p to within its normal range in primary hyperparathyroidism. In addition, the model provides a simulation of CYP24A1 inactivation that leads to a situation reminiscent of infantile hypercalcemia. In summary, our model of calcium handling can be used to decipher the complex regulation of calcium homeostasis.
Collapse
Affiliation(s)
- David Granjon
- Sorbonne Universités, UPMC Univ Paris 06, Université Paris Descartes, Sorbonne Paris Cité, INSERM UMRS 1138, CNRS ERL 8228, Centre de Recherche des Cordeliers, Paris, France; and.,Department of Pharmacology and Toxicology, University of Lausanne, and Service of Nephrology, Lausanne University Hospital, Lausanne, Switzerland
| | - Olivier Bonny
- Department of Pharmacology and Toxicology, University of Lausanne, and Service of Nephrology, Lausanne University Hospital, Lausanne, Switzerland
| | - Aurélie Edwards
- Sorbonne Universités, UPMC Univ Paris 06, Université Paris Descartes, Sorbonne Paris Cité, INSERM UMRS 1138, CNRS ERL 8228, Centre de Recherche des Cordeliers, Paris, France; and
| |
Collapse
|
60
|
Briffa JF, Grinfeld E, Jenkin KA, Mathai ML, Poronnik P, McAinch AJ, Hryciw DH. Diet induced obesity in rats reduces NHE3 and Na(+) /K(+) -ATPase expression in the kidney. Clin Exp Pharmacol Physiol 2016; 42:1118-26. [PMID: 26173747 DOI: 10.1111/1440-1681.12452] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 06/25/2015] [Accepted: 07/01/2015] [Indexed: 01/25/2023]
Abstract
The consumption of a high fat diet (HFD) is associated with proteinuria and altered sodium handling and excretion, which can lead to kidney disease. In the proximal tubule, the Na(+) /H(+) Exchanger 3 (NHE3) is responsible for normal protein reabsorption and the reabsorption of approximately 70% of the renal sodium load. It is the Na(+) /K(+) -ATPase that provides the driving force for the reabsorption of sodium and its exit across the basolateral membrane. This study investigates the effects that consumption of a HFD for 12 weeks has on NHE3 and Na(+) /K(+) -ATPase expression in the kidney. Western blot analysis identified a significant reduction in NHE3 and its modulator, phosphorylated protein kinase B, in renal lysate from obese rats. In the obese rats, a reduction in NHE3 expression in the proximal tubule may impact on the acidification of endosomes which are responsible for albumin uptake, suggesting a key role for the exchanger in protein endocytosis in obesity. Western blot analysis identified a reduction in Na(+) /K(+) -ATPase which could also potentially impact on albumin uptake and sodium reabsorption. This study demonstrates that consumption of a HFD for 12 weeks reduces renal NHE3 and Na(+) /K(+) -ATPase expression, an effect that may contribute to the albuminuria associated with obesity. Furthermore the reduction in these transporters is not likely to contribute to the reduced sodium excretion in obesity. These data highlight a potential link between NHE3 and Na(+) /K(+) -ATPase in the pathophysiological changes in renal protein handling observed in obesity.
Collapse
Affiliation(s)
- J F Briffa
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, St Albans, Victoria, Australia.,Department of Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - E Grinfeld
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, St Albans, Victoria, Australia
| | - K A Jenkin
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, St Albans, Victoria, Australia
| | - M L Mathai
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, St Albans, Victoria, Australia
| | - P Poronnik
- Department of Physiology, School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - A J McAinch
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, St Albans, Victoria, Australia
| | - D H Hryciw
- Department of Physiology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
61
|
Moor MB, Bonny O. Ways of calcium reabsorption in the kidney. Am J Physiol Renal Physiol 2016; 310:F1337-50. [PMID: 27009338 DOI: 10.1152/ajprenal.00273.2015] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 03/17/2016] [Indexed: 11/22/2022] Open
Abstract
The role of the kidney in calcium homeostasis has been reshaped from a classic view in which the kidney was regulated by systemic calcitropic hormones such as vitamin D3 or parathyroid hormone to an organ actively taking part in the regulation of calcium handling. With the identification of the intrinsic renal calcium-sensing receptor feedback system, the regulation of paracellular calcium transport involving claudins, and new paracrine regulators such as klotho, the kidney has emerged as a crucial modulator not only of calciuria but also of calcium homeostasis. This review summarizes recent molecular and endocrine contributors to renal calcium handling and highlights the tight link between calcium and sodium reabsorption in the kidney.
Collapse
Affiliation(s)
- Matthias B Moor
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland; and
| | - Olivier Bonny
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland; and Service of Nephrology, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
62
|
Huang HS, Ma MC. High Sodium-Induced Oxidative Stress and Poor Anticrystallization Defense Aggravate Calcium Oxalate Crystal Formation in Rat Hyperoxaluric Kidneys. PLoS One 2015; 10:e0134764. [PMID: 26241473 PMCID: PMC4524621 DOI: 10.1371/journal.pone.0134764] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 07/13/2015] [Indexed: 11/26/2022] Open
Abstract
Enhanced sodium excretion is associated with intrarenal oxidative stress. The present study evaluated whether oxidative stress caused by high sodium (HS) may be involved in calcium oxalate crystal formation. Male rats were fed a sodium-depleted diet. Normal-sodium and HS diets were achieved by providing drinking water containing 0.3% and 3% NaCl, respectively. Rats were fed a sodium-depleted diet with 5% hydroxyl-L-proline (HP) for 7 and 42 days to induce hyperoxaluria and/or calcium oxalate deposition. Compared to normal sodium, HS slightly increased calcium excretion despite diuresis; however, the result did not reach statistical significance. HS did not affect the hyperoxaluria, hypocalciuria or supersaturation caused by HP; however, it increased calcium oxalate crystal deposition soon after 7 days of co-treatment. Massive calcium oxalate formation and calcium crystal excretion in HS+HP rats were seen after 42 days of treatment. HP-mediated hypocitraturia was further exacerbated by HS. Moreover, HS aggravated HP-induced renal injury and tubular damage via increased apoptosis and oxidative stress. Increased urinary malondialdehyde excretion, in situ superoxide production, NAD(P)H oxidase and xanthine oxidase expression and activity, and decreased antioxidant enzyme expression or activity in the HS+HP kidney indicated exaggerated oxidative stress. Interestingly, this redox imbalance was associated with reduced renal osteopontin and Tamm-Horsfall protein expression (via increased excretion) and sodium-dependent dicarboxylate cotransporter NaDC-1 upregulation. Collectively, our results demonstrate that a HS diet induces massive crystal formation in the hyperoxaluric kidney; this is not due to increased urinary calcium excretion but is related to oxidative injury and loss of anticrystallization defense.
Collapse
Affiliation(s)
- Ho-Shiang Huang
- Department of Urology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Ming-Chieh Ma
- School of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
- * E-mail:
| |
Collapse
|
63
|
Alexander RT, Beggs MR, Zamani R, Marcussen N, Frische S, Dimke H. Ultrastructural and immunohistochemical localization of plasma membrane Ca2+-ATPase 4 in Ca2+-transporting epithelia. Am J Physiol Renal Physiol 2015; 309:F604-16. [PMID: 26180241 DOI: 10.1152/ajprenal.00651.2014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 07/11/2015] [Indexed: 01/07/2023] Open
Abstract
Plasma membrane Ca(2+)-ATPases (PMCAs) participate in epithelial Ca(2+) transport and intracellular Ca(2+) signaling. The Pmca4 isoform is enriched in distal nephron isolates and decreased in mice lacking the epithelial transient receptor potential vanilloid 5 Ca(2+) channel. We therefore hypothesized that Pmca4 plays a significant role in transcellular Ca(2+) flux and investigated the localization and regulation of Pmca4 in Ca(2+)-transporting epithelia. Using antibodies directed specifically against Pmca4, we found it expressed only in the smooth muscle layer of mouse and human intestines, whereas pan-specific Pmca antibodies detected Pmca1 in lateral membranes of enterocytes. In the kidney, Pmca4 showed broad localization to the distal nephron. In the mouse, expression was most abundant in segments coexpressing the epithelial ransient receptor potential vanilloid 5 Ca(2+) channel. Significant, albeit lower, expression was also evident in the region encompassing the cortical thick ascending limbs, macula densa, and early distal tubules as well as smooth muscle layers surrounding renal vessels. In the human kidney, a similar pattern of distribution was observed, with the highest PMCA4 expression in Na(+)-Cl(-) cotransporter-positive tubules. Electron microscopy demonstrated Pmca4 localization in distal nephron cells at both the basolateral membrane and intracellular perinuclear compartments but not submembranous vesicles, suggesting rapid trafficking to the plasma membrane is unlikely to occur in vivo. Pmca4 expression was not altered by perturbations in Ca(2+) balance, pointing to a housekeeping function of the pump in Ca(2+)-transporting epithelia. In conclusion, Pmca4 shows a divergent expression pattern in Ca(2+)-transporting epithelia, inferring diverse roles for this isoform not limited to transepithelial Ca(2+) transport.
Collapse
Affiliation(s)
- R Todd Alexander
- Department of Pediatrics, The University of Alberta, Edmonton, Alberta, Canada; Membrane Protein Disease Research Group, The University of Alberta, Edmonton, Alberta, Canada
| | - Megan R Beggs
- Membrane Protein Disease Research Group, The University of Alberta, Edmonton, Alberta, Canada
| | - Reza Zamani
- Department of Urology, Odense University Hospital, Odense, Denmark
| | - Niels Marcussen
- Department of Clinical Pathology, Odense University Hospital, Odense, Denmark
| | - Sebastian Frische
- Department of Biomedicine, University of Aarhus, Aarhus, Denmark; and
| | - Henrik Dimke
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Demark
| |
Collapse
|
64
|
Krishnan D, Liu L, Wiebe SA, Casey JR, Cordat E, Alexander RT. Carbonic anhydrase II binds to and increases the activity of the epithelial sodium-proton exchanger, NHE3. Am J Physiol Renal Physiol 2015; 309:F383-92. [PMID: 26041446 DOI: 10.1152/ajprenal.00464.2014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 05/20/2015] [Indexed: 02/08/2023] Open
Abstract
Two-thirds of sodium filtered by the renal glomerulus is reabsorbed from the proximal tubule via a sodium/proton exchanger isoform 3 (NHE3)-dependent mechanism. Since sodium and bicarbonate reabsorption are coupled, we postulated that the molecules involved in their reabsorption [NHE3 and carbonic anhydrase II (CAII)] might physically and functionally interact. Consistent with this, CAII and NHE3 were closely associated in a renal proximal tubular cell culture model as revealed by a proximity ligation assay. Direct physical interaction was confirmed in solid-phase binding assays with immobilized CAII and C-terminal NHE3 glutathione-S-transferase fusion constructs. To assess the effect of CAII on NHE3 function, we expressed NHE3 in a proximal tubule cell line and measured NHE3 activity as the rate of intracellular pH recovery, following an acid load. NHE3-expressing cells had a significantly greater rate of intracellular pH recovery than controls. Inhibition of endogenous CAII activity with acetazolamide significantly decreased NHE3 activity, indicating that CAII activates NHE3. To ascertain whether CAII binding per se activates NHE3, we expressed NHE3 with wild-type CAII, a catalytically inactive CAII mutant (CAII-V143Y), or a mutant unable to bind other transporters (CAII-HEX). NHE3 activity increased upon wild-type CAII coexpression, but not in the presence of the CAII V143Y or HEX mutant. Together these studies support an association between CAII and NHE3 that alters the transporter's activity.
Collapse
Affiliation(s)
- Devishree Krishnan
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada; and
| | - Lei Liu
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Shane A Wiebe
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada; and
| | - Joseph R Casey
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada; and
| | - Emmanuelle Cordat
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada; and
| | - R Todd Alexander
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada; and Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
65
|
Fenton RA, Poulsen SB, de la Mora Chavez S, Soleimani M, Busslinger M, Dominguez Rieg JA, Rieg T. Caffeine-induced diuresis and natriuresis is independent of renal tubular NHE3. Am J Physiol Renal Physiol 2015; 308:F1409-20. [PMID: 25925253 DOI: 10.1152/ajprenal.00129.2015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 04/18/2015] [Indexed: 01/15/2023] Open
Abstract
Caffeine is one of the most widely consumed behavioral substances. We have previously shown that caffeine- and theophylline-induced inhibition of renal reabsorption causes diuresis and natriuresis, an effect that requires functional adenosine A1 receptors. In this study, we tested the hypothesis that blocking the Gi protein-coupled adenosine A1 receptor via the nonselective adenosine receptor antagonist caffeine changes Na(+)/H(+) exchanger isoform 3 (NHE3) localization and phosphorylation, resulting in diuresis and natriuresis. We generated tubulus-specific NHE3 knockout mice (Pax8-Cre), where NHE3 abundance in the S1, S2, and S3 segments of the proximal tubule was completely absent or severely reduced (>85%) in the thick ascending limb. Consumption of fluid and food, as well as glomerular filtration rate, were comparable in control or tubulus-specific NHE3 knockout mice under basal conditions, while urinary pH was significantly more alkaline without evidence for metabolic acidosis. Caffeine self-administration increased total fluid and food intake comparably between genotypes, without significant differences in consumption of caffeinated solution. Acute caffeine application via oral gavage elicited a diuresis and natriuresis that was comparable between control and tubulus-specific NHE3 knockout mice. The diuretic and natriuretic response was independent of changes in total NHE3 expression, phosphorylation of serine-552 and serine-605, or apical plasma membrane NHE3 localization. Although caffeine had no clear effect on localization of the basolateral Na(+)/bicarbonate cotransporter NBCe1, pretreatment with DIDS inhibited caffeine-induced diuresis and natriuresis. In summary, NHE3 is not required for caffeine-induced diuresis and natriuresis.
Collapse
Affiliation(s)
- Robert A Fenton
- InterPrET Center, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Søren B Poulsen
- InterPrET Center, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Manoocher Soleimani
- Department of Medicine, University of Cincinnati, Cincinnati, Ohio; Research Services, Veterans Affairs Medical Center, Cincinnati, Ohio
| | | | - Jessica A Dominguez Rieg
- VA San Diego Healthcare System, San Diego, California; Bastyr University California, San Diego, California; and
| | - Timo Rieg
- VA San Diego Healthcare System, San Diego, California; Department of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
66
|
Vilas G, Krishnan D, Loganathan SK, Malhotra D, Liu L, Beggs MR, Gena P, Calamita G, Jung M, Zimmermann R, Tamma G, Casey JR, Alexander RT. Increased water flux induced by an aquaporin-1/carbonic anhydrase II interaction. Mol Biol Cell 2015; 26:1106-18. [PMID: 25609088 PMCID: PMC4357510 DOI: 10.1091/mbc.e14-03-0812] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aquaporin-1 (AQP1) enables greatly enhanced water flux across plasma membranes. The cytosolic carboxy terminus of AQP1 has two acidic motifs homologous to known carbonic anhydrase II (CAII) binding sequences. CAII colocalizes with AQP1 in the renal proximal tubule. Expression of AQP1 with CAII in Xenopus oocytes or mammalian cells increased water flux relative to AQP1 expression alone. This required the amino-terminal sequence of CAII, a region that binds other transport proteins. Expression of catalytically inactive CAII failed to increase water flux through AQP1. Proximity ligation assays revealed close association of CAII and AQP1, an effect requiring the second acidic cluster of AQP1. This motif was also necessary for CAII to increase AQP1-mediated water flux. Red blood cell ghosts resealed with CAII demonstrated increased osmotic water permeability compared with ghosts resealed with albumin. Water flux across renal cortical membrane vesicles, measured by stopped-flow light scattering, was reduced in CAII-deficient mice compared with wild-type mice. These data are consistent with CAII increasing water conductance through AQP1 by a physical interaction between the two proteins.
Collapse
Affiliation(s)
- Gonzalo Vilas
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Devishree Krishnan
- Department of Physiology, University of Alberta, Edmonton, AB T6G 1C9, Canada Membrane Protein Disease Research Group, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Sampath Kumar Loganathan
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 1C9, Canada Membrane Protein Disease Research Group, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Darpan Malhotra
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 1C9, Canada Membrane Protein Disease Research Group, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Lei Liu
- Department of Physiology, University of Alberta, Edmonton, AB T6G 1C9, Canada Membrane Protein Disease Research Group, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Megan Rachele Beggs
- Department of Physiology, University of Alberta, Edmonton, AB T6G 1C9, Canada Membrane Protein Disease Research Group, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Patrizia Gena
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70121 Bari, Italy
| | - Giuseppe Calamita
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70121 Bari, Italy
| | - Martin Jung
- Department of Medical Biochemistry and Molecular Biology, Saarland University, D-66424 Homburg, Germany
| | - Richard Zimmermann
- Department of Medical Biochemistry and Molecular Biology, Saarland University, D-66424 Homburg, Germany
| | - Grazia Tamma
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70121 Bari, Italy
| | - Joseph Roman Casey
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 1C9, Canada Department of Physiology, University of Alberta, Edmonton, AB T6G 1C9, Canada Membrane Protein Disease Research Group, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Robert Todd Alexander
- Department of Physiology, University of Alberta, Edmonton, AB T6G 1C9, Canada Membrane Protein Disease Research Group, University of Alberta, Edmonton, AB T6G 1C9, Canada Department of Pediatrics, University of Alberta, Edmonton, AB T6G 1C9, Canada
| |
Collapse
|
67
|
Alexander RT, Rievaj J, Dimke H. Paracellular calcium transport across renal and intestinal epithelia. Biochem Cell Biol 2014; 92:467-80. [PMID: 25386841 DOI: 10.1139/bcb-2014-0061] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Calcium (Ca(2+)) is a key constituent in a myriad of physiological processes from intracellular signalling to the mineralization of bone. As a consequence, Ca(2+) is maintained within narrow limits when circulating in plasma. This is accomplished via regulated interplay between intestinal absorption, renal tubular reabsorption, and exchange with bone. Many studies have focused on the highly regulated active transcellular transport pathways for Ca(2+) from the duodenum of the intestine and the distal nephron of the kidney. However, comparatively little work has examined the molecular constituents creating the paracellular shunt across intestinal and renal epithelium, the transport pathway responsible for the majority of transepithelial Ca(2+) flux. More specifically, passive paracellular Ca(2+) absorption occurs across the majority of the intestine in addition to the renal proximal tubule and thick ascending limb of Henle's loop. Importantly, recent studies demonstrated that Ca(2+) transport through the paracellular shunt is significantly regulated. Therefore, we have summarized the evidence for different modes of paracellular Ca(2+) flux across renal and intestinal epithelia and highlighted recent molecular insights into both the mechanism of secondarily active paracellular Ca(2+) movement and the identity of claudins that permit the passage of Ca(2+) through the tight junction of these epithelia.
Collapse
Affiliation(s)
- R Todd Alexander
- a Department of Pediatrics, The University of Alberta, 4-585 Edmonton Clinic Health Academy, 11405 - 87 Ave, Edmonton, AB T6G 2R7, Canada
| | | | | |
Collapse
|
68
|
Longo-Silva G, Toloni MHDA, de Menezes RCE, Temteo TL, Oliveira MAA, Asakura L, Costa EC, Taddei JADAC. Intake of protein, calcium and sodium in public child day care centers. REVISTA PAULISTA DE PEDIATRIA 2014; 32:193-9. [PMID: 25119750 PMCID: PMC4183027 DOI: 10.1590/0103-0582201432214613] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 11/25/2013] [Indexed: 11/21/2022]
Abstract
OBJECTIVE: To assess calcium, protein and sodium intake, of children that attend public
day-care centers and to compare it with the recommended one. METHODS: Cross-sectional descriptive study in seven public day care centers of São Paulo
city, Southeast Brazil, which enrolled 366 children between 12 and 36 months of
age. The data collection occurred between September and December 2010. Each day
care center was evaluated for three non-consecutive days, totaling 42 days and 210
meals. Dietary intake was assessed by a direct food weighing method. For the
nutritional calculation, DietWin(r) Profissional 2.0 was used, and the
adequacy was calculated according to the recommendations of the National School
Feeding Program for energy, protein, calcium and sodium. The calcium/protein
relation was also calculated, as well as calcium density (mg/1,000kcal). RESULTS: The energy (406.4kcal), protein (18.2g) and calcium (207.6mg) consumption did not
reach the recommended values in all the evaluated day care centers. Sodium
intake exceeded up to three times the recommendation. The calcium/protein ratio of
11.7mg/g was less than the adequate one (20mg/g). CONCLUSIONS: There was inadequacy of calcium, protein and sodium dietary intake, in children
attending public day-care centers.
Collapse
Affiliation(s)
| | | | | | | | | | - Leiko Asakura
- Faculdade de Nutrição, Universidade Federal de Alagoas, Maceió, AL, Brasil
| | | | | |
Collapse
|
69
|
De S, Kuwahara S, Saito A. The endocytic receptor megalin and its associated proteins in proximal tubule epithelial cells. MEMBRANES 2014; 4:333-55. [PMID: 25019425 PMCID: PMC4194038 DOI: 10.3390/membranes4030333] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 06/23/2014] [Accepted: 07/01/2014] [Indexed: 02/08/2023]
Abstract
Receptor-mediated endocytosis in renal proximal tubule epithelial cells (PTECs) is important for the reabsorption and metabolization of proteins and other substances, including carrier-bound vitamins and trace elements, in glomerular filtrates. Impairment of this endocytic process results in the loss of such substances and development of proteinuria, which is an important clinical indicator of kidney diseases and is also a risk marker for cardiovascular disease. Megalin, a member of the low-density lipoprotein receptor gene family, is a multiligand receptor expressed in the apical membrane of PTECs and plays a central role in the endocytic process. Megalin interacts with various intracellular adaptor proteins for intracellular trafficking and cooperatively functions with other membrane molecules, including the cubilin-amnionless complex. Evidence suggests that megalin and the cubilin-amnionless complex are involved in the uptake of toxic substances into PTECs, which leads to the development of kidney disease. Studies of megalin and its associated molecules will be useful for future development of novel strategies for the diagnosis and treatment of kidney diseases.
Collapse
Affiliation(s)
- Shankhajit De
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata 951-8510, Japan.
| | - Shoji Kuwahara
- Department of Applied Molecular Medicine, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata 951-8510, Japan.
| | - Akihiko Saito
- Department of Applied Molecular Medicine, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata 951-8510, Japan.
| |
Collapse
|
70
|
Abstract
Capasso et al. show a role of the calcium-sensing receptor (CaSR) in enhancing proximal tubular fluid absorption and urinary acidification by stimulation of luminal Na(+)/H(+) exchanger (NHE) activity. NHE3 is required for sodium and fluid absorption, and its activity is coupled to passive reabsorption of a major fraction of calcium through the paracellular route. These data shed new light on the regulation of the kidney by the CaSR and whether it directly affects proximal tubular functions.
Collapse
|
71
|
Fuster DG, Alexander RT. Traditional and emerging roles for the SLC9 Na+/H+ exchangers. Pflugers Arch 2013; 466:61-76. [PMID: 24337822 DOI: 10.1007/s00424-013-1408-8] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 11/14/2013] [Accepted: 11/20/2013] [Indexed: 10/25/2022]
Abstract
The SLC9 gene family encodes Na(+)/H(+) exchangers (NHEs). These transmembrane proteins transport ions across lipid bilayers in a diverse array of species from prokaryotes to eukaryotes, including plants, fungi, and animals. They utilize the electrochemical gradient of one ion to transport another ion against its electrochemical gradient. Currently, 13 evolutionarily conserved NHE isoforms are known in mammals [22, 46, 128]. The SLC9 gene family (solute carrier classification of transporters: www.bioparadigms.org) is divided into three subgroups [46]. The SLC9A subgroup encompasses plasmalemmal isoforms NHE1-5 (SLC9A1-5) and the predominantly intracellular isoforms NHE6-9 (SLC9A6-9). The SLC9B subgroup consists of two recently cloned isoforms, NHA1 and NHA2 (SLC9B1 and SLC9B2, respectively). The SLC9C subgroup consist of a sperm specific plasmalemmal NHE (SLC9C1) and a putative NHE, SLC9C2, for which there is currently no functional data [46]. NHEs participate in the regulation of cytosolic and organellar pH as well as cell volume. In the intestine and kidney, NHEs are critical for transepithelial movement of Na(+) and HCO3(-) and thus for whole body volume and acid-base homeostasis [46]. Mutations in the NHE6 or NHE9 genes cause neurological disease in humans and are currently the only NHEs directly linked to human disease. However, it is becoming increasingly apparent that members of this gene family contribute to the pathophysiology of multiple human diseases.
Collapse
Affiliation(s)
- Daniel G Fuster
- Division of Nephrology, Hypertension and Clinical Pharmacology and Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland,
| | | |
Collapse
|
72
|
van Angelen AA, San-Cristobal P, Pulskens WP, Hoenderop JG, Bindels RJ. The impact of dietary magnesium restriction on magnesiotropic and calciotropic genes. Nephrol Dial Transplant 2013; 28:2983-93. [DOI: 10.1093/ndt/gft358] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
73
|
Abegg K, Gehring N, Wagner CA, Liesegang A, Schiesser M, Bueter M, Lutz TA. Roux-en-Y gastric bypass surgery reduces bone mineral density and induces metabolic acidosis in rats. Am J Physiol Regul Integr Comp Physiol 2013; 305:R999-R1009. [PMID: 24026074 DOI: 10.1152/ajpregu.00038.2013] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Roux-en-Y gastric bypass (RYGB) surgery leads to bone loss in humans, which may be caused by vitamin D and calcium malabsorption and subsequent secondary hyperparathyroidism. However, because these conditions occur frequently in obese people, it is unclear whether they are the primary causes of bone loss after RYGB. To determine the contribution of calcium and vitamin D malabsorption to bone loss in a rat RYGB model, adult male Wistar rats were randomized for RYGB surgery, sham-operation-ad libitum fed, or sham-operation-body weight-matched. Bone mineral density, calcium and phosphorus balance, acid-base status, and markers of bone turnover were assessed at different time points for 14 wk after surgery. Bone mineral density decreased for several weeks after RYGB. Intestinal calcium absorption was reduced early after surgery, but plasma calcium and parathyroid hormone levels were normal. 25-hydroxyvitamin D levels decreased, while levels of active 1,25-dihydroxyvitamin D increased after surgery. RYGB rats displayed metabolic acidosis due to increased plasma lactate levels and increased urinary calcium loss throughout the study. These results suggest that initial calcium malabsorption may play a key role in bone loss early after RYGB in rats, but other factors, including chronic metabolic acidosis, contribute to insufficient bone restoration after normalization of intestinal calcium absorption. Secondary hyperparathyroidism is not involved in postoperative bone loss. Upregulated vitamin D activation may compensate for any vitamin D malabsorption.
Collapse
Affiliation(s)
- Kathrin Abegg
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Zurich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
74
|
Extraintestinal manifestations in an infant with microvillus inclusion disease: complications or features of the disease? Eur J Pediatr 2013; 172:1271-5. [PMID: 23354788 DOI: 10.1007/s00431-013-1948-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 01/14/2013] [Indexed: 01/23/2023]
Abstract
UNLABELLED Microvillus inclusion disease (MVID), a rare severe congenital enteropathy characterized by intracytoplasmic microvillous inclusions and variable brush border atrophy on intestinal epithelial cells histology, is associated with defective synthesis or abnormal function of the motor protein myosin Vb encoded by the MYO5B gene. Although MYO5B gene is expressed in all epithelial tissues, it is unclear so far whether organs other than intestine are affected in MVID patients. We report a case of an infant with MVID who presented liver dysfunction, hematuria, and Pneumocystis jiroveci pneumonia during the course of the disease. It is discussed whether extraintestinal manifestations in this patient are secondary consequences of MVID or might be features of the disease associated with altered MYO5B function. CONCLUSIONS MVID is classically included in the differential diagnosis of congenital diarrhea of secretory type. Recent advances in our knowledge regarding the role of myosin Vb in the pathophysiology of MVID is expected to clarify the clinical spectrum of the disease and the possible primary involvement of organs other than intestine.
Collapse
|
75
|
Rievaj J, Pan W, Cordat E, Alexander RT. The Na⁺/H⁺ exchanger isoform 3 is required for active paracellular and transcellular Ca²⁺ transport across murine cecum. Am J Physiol Gastrointest Liver Physiol 2013; 305:G303-13. [PMID: 23764894 PMCID: PMC4959879 DOI: 10.1152/ajpgi.00490.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal calcium (Ca²⁺) absorption occurs via paracellular and transcellular pathways. Although the transcellular route has been extensively studied, mechanisms mediating paracellular absorption are largely unexplored. Unlike passive diffusion, secondarily active paracellular Ca²⁺ uptake occurs against an electrochemical gradient with water flux providing the driving force. Water movement is dictated by concentration differences that are largely determined by Na⁺ fluxes. Consequently, we hypothesized that Na⁺ absorption mediates Ca²⁺ flux. NHE3 is central to intestinal Na⁺ absorption. NHE3 knockout mice (NHE3-/-) display impaired intestinal Na⁺, water, and Ca²⁺ absorption. However, the mechanism mediating this latter abnormality is not clear. To investigate this, we used Ussing chambers to measure net Ca²⁺ absorption across different segments of wild-type mouse intestine. The cecum was the only segment with net Ca²⁺ absorption. Quantitative RT-PCR measurements revealed cecal expression of all genes implicated in intestinal Ca²⁺ absorption, including NHE3. We therefore employed this segment for further studies. Inhibition of NHE3 with 100 μM 5-(N-ethyl-N-isopropyl) amiloride decreased luminal-to-serosal and increased serosal-to-luminal Ca²⁺ flux. NHE3-/- mice had a >60% decrease in luminal-to-serosal Ca²⁺ flux. Ussing chambers experiments under altered voltage clamps (-25, 0, +25 mV) showed decreased transcellular and secondarily active paracellular Ca²⁺ absorption in NHE3-/- mice relative to wild-type animals. Consistent with this, cecal Trpv6 expression was diminished in NHE3-/- mice. Together these results implicate NHE3 in intestinal Ca(2+) absorption and support the theory that this is, at least partially, due to the role of NHE3 in Na⁺ and water absorption.
Collapse
Affiliation(s)
- Juraj Rievaj
- Dept. of Pediatrics, 4-585 Edmonton Clinic Health Academy, 11405 87th Ave., Univ. of Alberta, Edmonton, Alberta, T6G 2R7, Canada.
| | | | | | | |
Collapse
|
76
|
Alexander RT, Dimke H, Cordat E. Proximal tubular NHEs: sodium, protons and calcium? Am J Physiol Renal Physiol 2013; 305:F229-36. [PMID: 23761670 DOI: 10.1152/ajprenal.00065.2013] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Na⁺/H⁺ exchange activity in the apical membrane of the proximal tubule is fundamental to the reabsorption of Na⁺ and water from the filtrate. The role of this exchange process in bicarbonate reclamation and, consequently, the maintenance of acid-base homeostasis has been appreciated for at least half a century and remains a pillar of renal tubular physiology. More recently, apical Na⁺/H⁺ exchange, mediated by Na⁺/H⁺ exchanger isoform 3 (NHE3), has been implicated in proximal tubular reabsorption of Ca²⁺ and Ca²⁺ homeostasis in general. Overexpression of NHE3 increased paracellular Ca²⁺ flux in a proximal tubular cell model. Consistent with this observation, mice with genetic deletion of Nhe3 have a noticable renal Ca²⁺ leak. These mice also display decreased intestinal Ca²⁺ uptake and osteopenia. This review highlights the traditional roles of proximal tubular Na⁺/H⁺ exchange and summarizes recent novel findings implicating the predominant isoform, NHE3, in Ca²⁺ homeostasis.
Collapse
Affiliation(s)
- R Todd Alexander
- Department of Pediatrics, Edmonton Clinic Health Academy, 11405-87 Ave., University of Alberta, Edmonton, AB, Canada T6G 2R7.
| | | | | |
Collapse
|
77
|
Lu Z, Ding L, Lu Q, Chen YH. Claudins in intestines: Distribution and functional significance in health and diseases. Tissue Barriers 2013; 1:e24978. [PMID: 24478939 PMCID: PMC3879173 DOI: 10.4161/tisb.24978] [Citation(s) in RCA: 189] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 05/06/2013] [Accepted: 05/08/2013] [Indexed: 02/08/2023] Open
Abstract
Intestines are organs that not only digest food and absorb nutrients, but also provide a defense barrier against pathogens and noxious agents ingested. Tight junctions (TJs) are the most apical component of the junctional complex, providing one form of cell-cell adhesion in enterocytes and playing a critical role in regulating paracellular barrier permeability. Alteration of TJs leads to a number of pathophysiological diseases causing malabsorption of nutrition and intestinal structure disruption, which may even contribute to systemic organ failure. Claudins are the major structural and functional components of TJs with at least 24 members in mammals. Claudins have distinct charge-selectivity, either by tightening the paracellular pathway or functioning as paracellular channels, regulating ions and small molecules passing through the paracellular pathway. In this review, we have discussed the functions of claudin family members, their distribution and localization in the intestinal tract of mammals, their alterations in intestine-related diseases and chemicals/agents that regulate the expression and localization of claudins as well as the intestinal permeability, which provide a therapeutic view for treating intestinal diseases.
Collapse
Affiliation(s)
- Zhe Lu
- Department of Basic Medicine; Hangzhou Normal University, Hangzhou, PR China ; Department of Anatomy and Cell Biology; Brody School of Medicine; East Carolina University; Greenville, NC USA
| | - Lei Ding
- Department of Anatomy and Cell Biology; Brody School of Medicine; East Carolina University; Greenville, NC USA ; Department of Oncology; Beijing Shijitan Hospital; Capital Medical University; Beijing, PR China
| | - Qun Lu
- Department of Anatomy and Cell Biology; Brody School of Medicine; East Carolina University; Greenville, NC USA
| | - Yan-Hua Chen
- Department of Anatomy and Cell Biology; Brody School of Medicine; East Carolina University; Greenville, NC USA
| |
Collapse
|
78
|
Dimke H, Desai P, Borovac J, Lau A, Pan W, Alexander RT. Activation of the Ca(2+)-sensing receptor increases renal claudin-14 expression and urinary Ca(2+) excretion. Am J Physiol Renal Physiol 2013; 304:F761-9. [PMID: 23283989 DOI: 10.1152/ajprenal.00263.2012] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Kidney stones are a prevalent clinical condition imposing a large economic burden on the healthcare system. Hypercalciuria remains the major risk factor for development of a Ca(2+)-containing stone. The kidney's ability to alter Ca(2+) excretion in response to changes in serum Ca(2+) is in part mediated by the Ca(2+)-sensing receptor (CaSR). Recent studies revealed renal claudin-14 (Cldn14) expression localized to the thick ascending limb (TAL) and its expression to be regulated via the CaSR. We find that Cldn14 expression is increased by high dietary Ca(2+) intake and by elevated serum Ca(2+) levels induced by prolonged 1,25-dihydroxyvitamin D3 administration. Consistent with this, activation of the CaSR in vivo via administration of the calcimimetic cinacalcet hydrochloride led to a 40-fold increase in Cldn14 mRNA. Moreover, overexpression of Cldn14 in two separate cell culture models decreased paracellular Ca(2+) flux by preferentially decreasing cation permeability, thereby increasing transepithelial resistance. These data support the existence of a mechanism whereby activation of the CaSR in the TAL increases Cldn14 expression, which in turn blocks the paracellular reabsorption of Ca(2+). This molecular mechanism likely facilitates renal Ca(2+) losses in response to elevated serum Ca(2+). Moreover, dysregulation of the newly described CaSR-Cldn14 axis likely contributes to the development of hypercalciuria and kidney stones.
Collapse
Affiliation(s)
- Henrik Dimke
- Department of Physiology and Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | |
Collapse
|
79
|
Yatabe MS, Yatabe J, Takano K, Murakami Y, Sakuta R, Abe S, Sanada H, Kimura J, Watanabe T. Effects of a high-sodium diet on renal tubule Ca2+ transporter and claudin expression in Wistar-Kyoto rats. BMC Nephrol 2012. [PMID: 23199000 PMCID: PMC3538060 DOI: 10.1186/1471-2369-13-160] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Urinary Ca2+ excretion increases with dietary NaCl. NaCl-induced calciuria may be associated with hypertension, urinary stone formation and osteoporosis, but its mechanism and long-term effects are not fully understood. This study examined alterations in the expressions of renal Ca2+ transporters, channels and claudins upon salt loading to better understand the mechanism of salt-induced urinary Ca2+ loss. Methods Eight-week old Wistar-Kyoto rats were fed either 0.3% or 8% NaCl diet for 8 weeks. Renal cortical expressions of Na+/Ca2+ exchanger 1 (NCX1), Ca2+ pump (PCMA1b), Ca2+ channel (TRPV5), calbindin-D28k, and claudins (CLDN-2, -7, -8, -16 and −19) were analyzed by quantitative PCR, western blot and/or immunohistochemistry. Results Fractional excretion of Ca2+ increased 6.0 fold with high-salt diet. Renal cortical claudin-2 protein decreased by approximately 20% with decreased immunological staining on tissue sections. Claudin-16 and −19 expressions were not altered. Renal cortical TRPV5, calbindin-D28k and NCX1 expressions increased 1.6, 1.5 and 1.2 fold, respectively. Conclusions Chronic high-salt diet decreased claudin-2 protein and increased renal TRPV5, calbindin-D28k, and NCX1. Salt loading is known to reduce the proximal tubular reabsorption of both Na+ and Ca2+. The reduction in claudin-2 protein expression may be partly responsible for the reduced Ca2+ reabsorption in this segment. The concerted upregulation of more distal Ca2+-transporting molecules may be a physiological response to curtail the loss of Ca2+, although the magnitude of compensation does not seem adequate to bring the urinary Ca2+ excretion down to that of the normal-diet group.
Collapse
Affiliation(s)
- Midori Sasaki Yatabe
- Department of Pharmacology, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Abstract
PURPOSE OF REVIEW In this review, we focus on the recent (March 2010 to September 2011) advances in small intestinal ion transport, with particular emphasis on sodium, chloride, bicarbonate, and calcium transport mechanisms under physiological and pathophysiological conditions. RECENT FINDINGS Knockout of NHERF1 and NHERF2 allowed translation of the data largely derived from the in-vitro models into a living organism. These studies also expand our knowledge about the complexity of intestinal transporter interactomes, define the role for scaffolding proteins in basal and regulated apical transport, and help identify potential targets for pharmacological approaches. We continue to accumulate novel information about the function and regulation of NHE3 (including its role in regulating paracellular Ca2+ flux), NHE8, as well as about the complexity of the intestinal Cl- and HCO3- transport in health and disease. SUMMARY Thanks to the new genetically engineered mouse models, a significant progress has been made in our understanding of the role of NHERF proteins in regulation of intestinal Na+ absorption. Significant novel data on the coordinated function of bicarbonate, chloride, and sodium transporters contributes to our current views of the integrative physiology of the small intestinal electrolyte transport.
Collapse
|