Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Cell Mol Life Sci ; 79(8): 418, 2022 Jul 12.
Article in English | MEDLINE | ID: mdl-35819535

ABSTRACT

Magnesium (Mg2+) is the most prevalent divalent intracellular cation. As co-factor in many enzymatic reactions, Mg2+ is essential for protein synthesis, energy production, and DNA stability. Disturbances in intracellular Mg2+ concentrations, therefore, unequivocally result in delayed cell growth and metabolic defects. To maintain physiological Mg2+ levels, all organisms rely on balanced Mg2+ influx and efflux via Mg2+ channels and transporters. This review compares the structure and the function of prokaryotic Mg2+ transporters and their eukaryotic counterparts. In prokaryotes, cellular Mg2+ homeostasis is orchestrated via the CorA, MgtA/B, MgtE, and CorB/C Mg2+ transporters. For CorA, MgtE, and CorB/C, the motifs that form the selectivity pore are conserved during evolution. These findings suggest that CNNM proteins, the vertebrate orthologues of CorB/C, also have Mg2+ transport capacity. Whereas CorA and CorB/C proteins share the gross quaternary structure and functional properties with their respective orthologues, the MgtE channel only shares the selectivity pore with SLC41 Na+/Mg2+ transporters. In eukaryotes, TRPM6 and TRPM7 Mg2+ channels provide an additional Mg2+ transport mechanism, consisting of a fusion of channel with a kinase. The unique features these TRP channels allow the integration of hormonal, cellular, and transcriptional regulatory pathways that determine their Mg2+ transport capacity. Our review demonstrates that understanding the structure and function of prokaryotic magnesiotropic proteins aids in our basic understanding of Mg2+ transport.


Subject(s)
Magnesium , Membrane Transport Proteins , Biological Transport , Cations, Divalent/metabolism , Magnesium/metabolism , Membrane Transport Proteins/metabolism , Phosphotransferases/metabolism
2.
BMC Med Educ ; 19(1): 97, 2019 Apr 03.
Article in English | MEDLINE | ID: mdl-30943962

ABSTRACT

BACKGROUND: A potential concern of formative testing using web-based applications ("apps") is provision of limited feedback. Adopting a randomised controlled trial in 463 first year (bio) medical students, we explored if providing immediate, detailed feedback during "app"-based formative testing can further improve study behaviour and study performance of (bio)medical students. METHODS: Students had access to a formative testing "app", which involved 7 formative test modules throughout the 4-week course. In a randomised order, subjects received the "app" with (n = 231, intervention) or without (n = 232, control) detailed feedback during the formative test modules. RESULTS: No differences in app-use was found between groups (P = 0.15), whereas the intervention group more frequently reviewed information compared to controls (P = 0.007). Exam scores differed between non-/moderate-/intensive- users of the "app" (P < 0.001). No differences in exam scores were found between intervention (6.6 ± 1.1) versus control (6.6 ± 1.1, P = 0.18). Time spent studying was significantly higher compared to previous courses in moderate- and intensive-users (P = 0.006 and < 0.001, respectively), but not in non-users (P = 0.55). Time spent studying did not differ between groups (P > 0.05). CONCLUSIONS: Providing detailed feedback did not further enhance the effect of a web-based application of formative testing on study behaviour or study performance in (bio)medical students, possibly because of a ceiling-effect.


Subject(s)
Education, Medical, Undergraduate , Formative Feedback , Mobile Applications/statistics & numerical data , Retention, Psychology/physiology , Smartphone/statistics & numerical data , Students, Medical , Computer-Assisted Instruction , Curriculum , Educational Measurement , Humans , Learning , Program Evaluation , Students, Medical/statistics & numerical data , Test Taking Skills
3.
Aliment Pharmacol Ther ; 43(11): 1178-85, 2016 06.
Article in English | MEDLINE | ID: mdl-27086738

ABSTRACT

BACKGROUND: Proton pump inhibitors (PPI) are among the most widely prescribed drugs to treat gastric acid-related disorders. PPI-induced hypomagnesaemia, a defect in intestinal absorption of Mg(2+) , can be a severe side effect of chronic PPI use. AIM: To restore serum Mg(2+) concentrations in PPI-induced hypomagnesaemia patients by dietary supplementation with inulin fibres. METHODS: Eleven patients with PPI-induced hypomagnesaemia and 10 controls were treated with inulin (20 g/day). Each trial consisted of two cycles of 14-day inulin treatment followed by a washout period of 14 days. Patients continued to use their PPI. Serum Mg(2+) levels served as the primary endpoint. RESULTS: Inulin significantly enhanced serum Mg(2+) levels from 0.60 to 0.68 mmol/L in PPI-induced hypomagnesaemia patients, and from 0.84 to 0.93 mmol/L in controls. As a consequence 24 h urinary Mg(2+) excretion was significantly increased in patients with PPI-induced hypomagnesaemia (0.3-2.2 mmol/day). Symptoms related to hypomagnesaemia, including muscle cramps and paraesthesia, were reduced during intervention with inulin. CONCLUSION: Inulin increases serum Mg(2+) concentrations under PPI maintenance in patients with PPI-induced hypomagnesaemia.


Subject(s)
Inulin/administration & dosage , Magnesium/blood , Proton Pump Inhibitors/adverse effects , Adult , Aged , Case-Control Studies , Female , Humans , Intestinal Absorption , Magnesium Deficiency/blood , Male , Middle Aged , Muscle Cramp/drug therapy , Proton Pump Inhibitors/therapeutic use , Young Adult
4.
Bone ; 57(2): 443-54, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24084385

ABSTRACT

We explored the role of transient receptor potential vanilloid 4 (TRPV4) in murine bone metabolism and association of TRPV4 gene variants with fractures in humans. Urinary and histomorphometrical analyses demonstrated reduced osteoclast activity and numbers in male Trpv4(-/-) mice, which was confirmed in bone marrow-derived osteoclast cultures. Osteoblasts and bone formation as shown by serum procollagen type 1 amino-terminal propeptide and histomorphometry, including osteoid surface, osteoblast and osteocyte numbers were not affected in vivo. Nevertheless, osteoblast differentiation was enhanced in Trpv4(-/-) bone marrow cultures. Cortical and trabecular bone mass was 20% increased in male Trpv4(-/-) mice, compared to sex-matched wild type (Trpv4(+/+)) mice. However, at the same time intracortical porosity was increased and bone matrix mineralization was reduced. Together, these lead to a maximum load, stiffness and work to failure of the femoral bone, which were not different compared to Trpv4(+/+) mice, while the bone material was less resistant to stress and less elastic. The differential impacts on these determinants of bone strength were likely responsible for the lack of any changes in whole bone strength in the Trpv4(-/-) mice. None of these skeletal parameters were affected in female Trpv4(-/-) mice. The T-allele of rs1861809 SNP in the TRPV4 locus was associated with a 30% increased risk (95% CI: 1.1-1.6; p=0.013) for non-vertebral fracture risk in men, but not in women, in the Rotterdam Study. Meta-analyses with the population-based LASA study confirmed the association with non-vertebral fractures in men. This was lost when the non-population-based studies Mr. OS and UFO were included. In conclusion, TRPV4 is a male-specific regulator of bone metabolism, a determinant of bone strength, and a potential risk predictor for fractures through regulation of bone matrix mineralization and intra-cortical porosity. This identifies TRPV4 as a unique sexually dimorphic therapeutic and/or diagnostic candidate for osteoporosis.


Subject(s)
Bone and Bones/pathology , Osteoporotic Fractures/epidemiology , Sex Characteristics , TRPV Cation Channels/deficiency , Animals , Bone and Bones/metabolism , Elastic Modulus , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Male , Mice , Netherlands/epidemiology , Osteoblasts/pathology , Osteoclasts/pathology , Osteoporotic Fractures/genetics , Phenotype , Polymorphism, Single Nucleotide/genetics , Risk Factors , Stress, Mechanical , TRPV Cation Channels/genetics
5.
Pflugers Arch ; 465(11): 1613-20, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23756852

ABSTRACT

Proton pump inhibitors (PPIs) are potent blockers of gastric acid secretion, used by millions of patients suffering from gastric acid-related complaints. Although PPIs have an excellent safety profile, an increasing number of case reports describe patients with severe hypomagnesemia due to long-term PPI use. As there is no evidence of a renal Mg²âº leak, PPI-induced hypomagnesemia is hypothesized to result from intestinal malabsorption of Mg²âº. The aim of this study was to investigate the effect of PPIs on Mg ²âºhomeostasis in an in vivo mouse model. To this end, C57BL/6J mice were treated with omeprazole, under normal and low dietary Mg²âº availability. Omeprazole did not induce changes in serum Mg²âº levels (1.48 ± 0.05 and 1.54 ± 0.05 mmol/L in omeprazole-treated and control mice, respectively), urinary Mg²âº excretion (35 ± 3 µmol/24 h and 30 ± 4 µmol/24 h in omeprazole-treated and control mice, respectively), or fecal Mg²âº excretion (84 ± 4 µmol/24 h and 76 ± 4 µmol/24 h in omeprazole-treated and control mice, respectively) under any of the tested experimental conditions. However, omeprazole treatment did increase the mRNA expression level of the transient receptor potential melastatin 6 (TRPM6), the predominant intestinal Mg²âº channel, in the colon (167 ± 15 and 100 ± 7 % in omeprazole-treated and control mice, respectively, P < 0.05). In addition, the expression of the colonic H⁺,K⁺-ATPase (cHK-α), a homolog of the gastric H⁺,K⁺-ATPase that is the primary target of omeprazole, was also significantly increased (354 ± 43 and 100 ± 24 % in omeprazole-treated and control mice, respectively, P < 0.05). The expression levels of other magnesiotropic genes remained unchanged. Based on these findings, we hypothesize that omeprazole inhibits cHK-α activity, resulting in reduced extrusion of protons into the large intestine. Since TRPM6-mediated Mg²âºabsorption is stimulated by extracellular protons, this would diminish the rate of intestinal Mg²âº absorption. The increase of TRPM6 expression in the colon may compensate for the reduced TRPM6 currents, thereby normalizing intestinal Mg²âº absorption during omeprazole treatment in C57BL/6J mice, explaining unchanged serum, urine, and fecal Mg²âº levels.


Subject(s)
Colon/metabolism , Omeprazole/pharmacology , Proton Pump Inhibitors/pharmacology , TRPM Cation Channels/metabolism , Animals , Colon/drug effects , H(+)-K(+)-Exchanging ATPase/genetics , H(+)-K(+)-Exchanging ATPase/metabolism , Homeostasis , Intestinal Absorption/drug effects , Magnesium/blood , Magnesium/metabolism , Magnesium/urine , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , TRPM Cation Channels/genetics , Transcription, Genetic
6.
Aliment Pharmacol Ther ; 36(5): 405-13, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22762246

ABSTRACT

BACKGROUND: Proton pump inhibitors (PPIs) are a mainstay therapy for all gastric acid-related diseases. Clinical concerns arise from a small but growing number of case reports presenting PPI-induced hypomagnesaemia (PPIH) as a consequence of long-term PPI use. Current opinion is that reduced intestinal magnesium absorption might be involved, but nothing is known on the molecular mechanism underlying PPIH. AIM: To investigate whether or not PPIH is a true, long-term drug-class effect of all PPIs and to scrutinise a possible role of comorbidity in its aetiology. Therefore, the primary objective in particular was to investigate serum magnesium dynamics in trials drug withdrawal and re-challenge. The secondary objective was to profile the 'patient at risk'. METHODS: We reviewed systematically all currently available case reports on the subject and performed a statistical analysis on extracted data. RESULTS: Proton pump inhibitor-induced hypomagnesaemia PPIH is a drug-class effect and occurred after 5.5 years (median) of PPI use, onset was broad and ranged from 14 days to 13 years. Discontinuation of PPIs resulted in fast recovery from PPIH in 4 days and re-challenge led to reoccurrence within 4 days. Histamine-2-receptor antagonists were the preferable replacement therapy in PPIH and prevented reoccurrence of hypomagnesaemia. In PPIH no specific risk profile was identified that was linked to the hypomagnesaemia. CONCLUSIONS: The cases of PPIH show severe symptoms of magnesium depletion and identification of its causation was only possible through withdrawal of the PPI. Clinical awareness of PPIH is key to avoid putting patients at risk.


Subject(s)
Gastrointestinal Diseases/drug therapy , Magnesium Deficiency/chemically induced , Magnesium/blood , Proton Pump Inhibitors/adverse effects , Clinical Trials as Topic , Humans , Magnesium/metabolism , Magnesium Deficiency/blood , Risk Factors
7.
Kidney Int ; 72(6): 663-5, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17805315

ABSTRACT

Controlling the development of vascular calcification in chronic kidney disease is essential, because it is associated with increased cardiovascular pathology. However, the precise mechanism of vascular calcification has not been completely elucidated. In the literature, the involvement of passive calcium and phosphate deposition as well as an active process stimulating the transformation of vascular smooth muscle cells into an osteoblastic phenotype is suggested. New promising insights into the etiology could lead to better treatment strategies, as Mizobuchi et al. now report.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Calcinosis/drug therapy , Ergocalciferols/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Animals , Calcinosis/pathology , Humans , Renal Insufficiency, Chronic/pathology
8.
Rev Physiol Biochem Pharmacol ; 158: 77-160, 2007.
Article in English | MEDLINE | ID: mdl-17729442

ABSTRACT

Ca2+ and Mg2+ are essential ions in a wide variety of cellular processes and form a major constituent of bone. It is, therefore, essential that the balance of these ions is strictly maintained. In the last decade, major breakthrough discoveries have vastly expanded our knowledge of the mechanisms underlying epithelial Ca2+ and Mg2+ transport. The genetic defects underlying various disorders with altered Ca2+ and/or Mg2+ handling have been determined. Recently, this yielded the molecular identification of TRPM6 as the gatekeeper of epithelial Mg2+ transport. Furthermore, expression cloning strategies have elucidated two novel members of the transient receptor potential family, TRPV5 and TRPV6, as pivotal ion channels determining transcellular Ca2+ transport. These two channels are regulated by a variety of factors, some historically strongly linked to Ca2+ homeostasis, others identified in a more serendipitous manner. Herein we review the processes of epithelial Ca2+ and Mg2+ transport, the molecular mechanisms involved, and the various forms of regulation.


Subject(s)
Calcium/metabolism , Epithelial Cells/physiology , Magnesium/metabolism , Animals , Biological Transport , Humans
9.
Handb Exp Pharmacol ; (179): 207-20, 2007.
Article in English | MEDLINE | ID: mdl-17217059

ABSTRACT

Ca2+ homeostasis in the body is tightly controlled, and is a balance between absorption in the intestine, excretion via the urine, and exchange from bone. Recently, the epithelial Ca2+ channel (TRPV5) has been identified as the gene responsible for the Ca2+ influx in epithelial cells of the renal distal convoluted tubule. TRPV5 is unique within the family of transient receptor potential (TRP) channels due to its high Ca2+ selectivity. Ca2+ flux through TRPV5 is controlled in three ways. First, TRPV5 gene expression is regulated by calciotropic hormones such as vitamin D3 and parathyroid hormone. Second, Ca2+ transport through TRPV5 is controlled by modulating channel activity. Intracellular Ca2+, for example, regulates channel activity by feedback inhibition. Third, TRPV5 is controlled by mobilization of the channel through trafficking toward the plasma membrane. The newly identified anti-aging hormone Klotho regulates TRPV5 by cleaving off sugar residues from the extracellular domain of the protein, resulting in a prolonged expression of TRPV5 at the plasma membrane. Inactivation of TRPV5 in mice leads to severe hypercalciuria, which is compensated by increased intestinal Ca2+ absorption due to augmented vitamin D3 levels. Furthermore, TRPV5 deficiency in mice is associated with polyuria, urine acidification, and reduced bone thickness. Some pharmaceutical compounds, such as the immunosuppressant FK506, affect the Ca2+ balance by modulating TRPV5 gene expression. This underlines the importance of elucidating the role of TRPV5 in Ca(2+)-related disorders, thereby enhancing the possibilities for pharmacological intervention. This chapter describes a unique TRP channel and highlights its regulation and function in renal Ca2+ reabsorption and overall Ca2+ homeostasis.


Subject(s)
Calcium Channels/genetics , Calcium Channels/physiology , Calcium/physiology , Homeostasis/physiology , TRPV Cation Channels/genetics , TRPV Cation Channels/physiology , Animals , Biotransformation/drug effects , Calcium Channels/drug effects , Electrophysiology , Humans , TRPV Cation Channels/drug effects
10.
Biochem Soc Trans ; 35(Pt 1): 115-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17233615

ABSTRACT

Ca(2+) is an essential ion in all organisms and many physiological functions in the body rely on the exact maintenance of the Ca(2+) balance. The epithelial Ca(2+) channels TRPV5 [TRP (transient receptor potential) vanilloid 5] and TRPV6 are the most Ca(2+)-selective members of the TRP superfamily and are generally considered as the gatekeepers of Ca(2+) entry across epithelia. TRPV5 is involved in Ca(2+) reabsorption from pro-urine, while TRPV6 has an essential role in intestinal Ca(2+) uptake. These channels are the prime targets of calciotropic hormonal regulation, including vitamin D and parathyroid hormone. In addition, extra- and intra-cellular signalling by associated proteins and Ca(2+) itself play key roles in TRPV5 and TRPV6 regulation. In this paper, we describe the present understanding of the concerted action of calbindin-D(28k), klotho and BSPRY (B-box and SPRY-domain-containing protein) at different levels throughout the epithelial cell to control Ca(2+) influx at the luminal entry gate.


Subject(s)
Calcium Channels/physiology , Gene Expression Regulation , TRPV Cation Channels/physiology , Animals , Calbindins , Calcium/metabolism , Cell Membrane/metabolism , Glucuronidase/metabolism , Humans , Ions , Klotho Proteins , Models, Biological , Parathyroid Hormone/metabolism , Protein Structure, Tertiary , S100 Calcium Binding Protein G/chemistry , Signal Transduction , Vitamin D/chemistry
11.
Kidney Int ; 69(4): 650-4, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16518325

ABSTRACT

Ca2+ homeostasis is an important factor, which is underlined by the numerous clinical symptoms that involve Ca2+ deficiencies. The overall Ca2+ balance is maintained by the concerted action of Ca2+ absorption in the intestine, reabsorption in the kidney, and exchange from bone, which are all under the control of the calciotropic hormones that are released upon a demand for Ca2+. In the kidney, these calciotropic hormones affect active Ca2+ reabsorption, which consists of TRPV5 as the apical entry gate for Ca2+ influx, calbindin-D28K as an intracellular ferry for Ca2+ and, NCX1 and PMCA1b for extrusion of Ca2+ across the basolateral membrane. This review highlights the action of hormones on renal Ca2+ handling and focuses on the coordinated control of the renal Ca2+ transport proteins. Parathyroid hormone stimulates renal Ca2+ handling by regulating active Ca2+ reabsorption on both the genomic and non-genomic level. Estrogens harbor calciotropic hormone characteristics positively regulating the expression of TRPV5, independently of vitamin D. Besides having a strong regulatory effect on the expression of the intestinal Ca2+ transport proteins, vitamin D contributes to the overall Ca2+ balance by enhancing the expression of the Ca2+ transport machinery in the kidney. Dietary Ca2+ is involved in regulating its own handling by controlling the expression of the renal Ca2+ transport proteins. Thus, the magnitude of Ca2+ entry via TRPV5 controls the expression of the other Ca2+ transport proteins underlining the gatekeeper function of this Ca2+ channel in the renal Ca2+ handling.


Subject(s)
Calcium/metabolism , Homeostasis , Kidney/metabolism , Absorption/drug effects , Animals , Calbindin 1 , Calbindins , Calcium/deficiency , Calcium Channels/physiology , Calcium, Dietary/pharmacology , Calcium-Transporting ATPases/physiology , Cation Transport Proteins/physiology , Estrogens/physiology , Humans , Parathyroid Hormone/physiology , Plasma Membrane Calcium-Transporting ATPases , S100 Calcium Binding Protein G/physiology , Sodium-Calcium Exchanger/physiology , TRPV Cation Channels/physiology , Vitamin D/physiology
12.
Science ; 310(5747): 490-3, 2005 Oct 21.
Article in English | MEDLINE | ID: mdl-16239475

ABSTRACT

Blood calcium concentration is maintained within a narrow range despite large variations in dietary input and body demand. The Transient Receptor Potential ion channel TRPV5 has been implicated in this process. We report here that TRPV5 is stimulated by the mammalian hormone klotho. Klotho, a beta-glucuronidase, hydrolyzes extracellular sugar residues on TRPV5, entrapping the channel in the plasma membrane. This maintains durable calcium channel activity and membrane calcium permeability in kidney. Thus, klotho activates a cell surface channel by hydrolysis of its extracellular N-linked oligosaccharides.


Subject(s)
Calcium Channels/metabolism , Membrane Proteins/metabolism , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Calcium Channels/genetics , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Glucuronidase/antagonists & inhibitors , Glucuronidase/metabolism , Glycosylation , Humans , Hydrolysis , Kidney/cytology , Kidney/metabolism , Klotho Proteins , Mice , Mice, Inbred C57BL , Mutation , Patch-Clamp Techniques , Protein Transport , Rabbits , Sodium/metabolism , TRPV Cation Channels/genetics , Transfection
13.
Ned Tijdschr Geneeskd ; 149(24): 1330-3, 2005 Jun 11.
Article in Dutch | MEDLINE | ID: mdl-16008036

ABSTRACT

Gitelman's syndrome is characterised by persistent hypokalaemia, hypomagnesaemia and hypocalciuria (OMIM 263800). This rare autosomal recessive disorder is caused by renal Na+, Cl-, K+ and Mg2+ wasting. Other typical features include hypocalciuria and an intact renal concentrating ability. Gitelman's syndrome is caused by mutations in the SLC12A3 gene, encoding the thiazide-sensitive sodium-chloride co-transporter (NCC). NCC is located in the distal convoluted tubule of the kidney, a segment known to play an important role in active magnesium reabsorption in the nephron. The exact mechanisms underlying hypomagnesaemia and hypocalciuria in Gitelman's syndrome are still poorly understood, but point to enhanced proximal Na+ and Ca2+ reabsorption and apoptosis of distal convoluted tubule cells.


Subject(s)
Calcium/metabolism , Chlorides/metabolism , Magnesium/metabolism , Receptors, Drug/genetics , Renal Tubular Transport, Inborn Errors/genetics , Sodium/metabolism , Symporters/genetics , Humans , Kidney Tubules, Distal/metabolism , Kidney Tubules, Distal/physiopathology , Renal Tubular Transport, Inborn Errors/physiopathology , Sodium Chloride Symporters , Solute Carrier Family 12, Member 3 , Syndrome
14.
EMBO J ; 22(4): 776-85, 2003 Feb 17.
Article in English | MEDLINE | ID: mdl-12574114

ABSTRACT

The molecular assembly of the epithelial Ca(2+) channels (TRPV5 and TRPV6) was investigated to determine the subunit stoichiometry and composition. Immunoblot analysis of Xenopus laevis oocytes expressing TRPV5 and TRPV6 revealed two specific bands of 75 and 85-100 kDa, corresponding to the core and glycosylated proteins, respectively, for each channel. Subsequently, membranes of these oocytes were sedimented on sucrose gradients. Immuno blotting revealed that TRPV5 and TRPV6 complexes migrate with a mol. wt of 400 kDa, in line with a tetrameric structure. The tetrameric stoichiometry was confirmed in an electrophysiological analysis of HEK293 cells co-expressing concatemeric channels together with a TRPV5 pore mutant that reduced Cd(2+) sensitivity and voltage-dependent gating. Immuno precipitations using membrane fractions from oocytes co-expressing TRPV5 and TRPV6 demonstrated that both channels can form heteromeric complexes. Expression of all possible heterotetrameric TRPV5/6 complexes in HEK293 cells resulted in Ca(2+) channels that varied with respect to Ca(2+)-dependent inactivation, Ba(2+) selectivity and pharmacological block. Thus, Ca(2+)-transporting epithelia co-expressing TRPV5 and TRPV6 can generate a pleiotropic set of functional heterotetrameric channels with different Ca(2+) transport kinetics.


Subject(s)
Calcium Channels/chemistry , Calcium/metabolism , Epithelium/metabolism , Animals , Calcium Channels/metabolism , Epithelium/chemistry , Kidney/chemistry , Kidney/metabolism , Mice , Precipitin Tests , Protein Structure, Tertiary , TRPV Cation Channels
15.
Cell Calcium ; 31(6): 253-64, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12098215

ABSTRACT

Calcium influx into the cell from the extracellular medium is crucial for important processes including muscle contraction, secretion and gene expression. This calcium influx is mainly mediated through calcium influx channels, which on the basis of their activation mechanism can be subdivided in voltage-gated calcium channels, which have already been thoroughly characterized and non-voltage-gated calcium permeable channels. This latter group includes ion channels activated by binding of extra and intracellular messengers, mechanical stress or depletion of intracellular calcium stores. Currently little molecular data is available concerning this class of calcium influx channels. However, recent studies have indicated that members of the transient receptor potential (TRP) family of ion channels can function as calcium influx channels both in excitable and non-excitable tissues. On the basis of structural information the TRP family is subdivided in three main subfamilies: the TRPC (canonical) group, the TRPV (vanilloid) group and the TRPM (melastatin) group. The cloning and characterization of members of this cation channel family has exploded during recent years, leading to a plethora of data concerning TRPs in a variety of tissues and species, including mammals, insects and yeast. This review summarizes the currently available information concerning members of the TRP family expressed in mammalian tissues.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Ion Channels/metabolism , Animals , Humans , Ion Channels/classification , Ion Channels/genetics , Multigene Family , Phylogeny , Protein Structure, Secondary , Tissue Distribution
16.
J Physiol ; 537(Pt 3): 747-61, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11744752

ABSTRACT

1. The epithelial Ca(2+) channel (ECaC) family represents a unique group of Ca(2+)-selective channels that share limited homology to the ligand-gated capsaicin receptors, the osmolarity-sensitive channel OTRPC4, as well as the transient receptor potential family. Southern blot analysis demonstrated that this family is restricted to two members, ECaC1 and ECaC2 (also named CaT1). 2. RT-PCR analysis demonstrated that the two channels are co-expressed in calbindin-D-containing epithelia, including small intestine, pancreas and placenta, whereas kidney and brain only express ECaC1 and stomach solely ECaC2. 3. From an electrophysiological point of view, ECaC1 and ECaC2 are highly similar channels. Differences concern divalent cation permeability, the kinetics of Ca(2+)-dependent inactivation and recovery from inactivation. 4. Ruthenium red is a potent blocker of ECaC activity. Interestingly, ECaC2 has a 100-fold lower affinity for ruthenium red (IC(50) 9 +/- 1 microM) than ECaC1 (IC(50) 121 +/- 13 nM). 5. ECaCs are modulated by intracellular Mg(2+) and ATP. ECaC1 and ECaC2 activity rapidly decay in the absence of intracellular ATP. This effect is further accelerated at higher intracellular Mg(2+) concentrations. 6. In conclusion, ECaC1 and ECaC2 are homologous channels, with an almost identical pore region. They can be discriminated by their sensitivity for ruthenium red and show differences in Ca(2+)-dependent regulation.


Subject(s)
Calcium Channels/physiology , Adenosine Triphosphate/physiology , Amino Acid Sequence/genetics , Animals , Calcium/pharmacology , Calcium/physiology , Calcium Channels/drug effects , Calcium Channels/genetics , Calcium Channels/metabolism , Cations, Divalent/metabolism , Cations, Monovalent/metabolism , Electric Conductivity , Magnesium/pharmacology , Magnesium/physiology , Mice , Molecular Sequence Data , Multigene Family/genetics , Phylogeny , Ruthenium Red/pharmacology , Sodium/pharmacology , TRPV Cation Channels , Tissue Distribution
17.
Am J Physiol Renal Physiol ; 281(6): F1021-7, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11704552

ABSTRACT

The organization of Na(+) and Ca(2+) transport pathways along the mouse distal nephron is incompletely known. We revealed by immunohistochemistry a set of Ca(2+) and Na(+) transport proteins along the mouse distal convolution. The thiazide-sensitive Na(+)-Cl(-) cotransporter (NCC) characterized the distal convoluted tubule (DCT). The amiloride-sensitive epithelial Na(+) channel (ENaC) colocalized with NCC in late DCT (DCT2) and extended to the downstream connecting tubule (CNT) and collecting duct (CD). In early DCT (DCT1), the basolateral Ca(2+)-extruding proteins [Na(+)/Ca(2+) exchanger (NCX), plasma membrane Ca(2+)-ATPase (PCMA)] and the cytoplasmic Ca(2+)-binding protein calbindin D(28K) (CB) were found at very low levels, whereas the cytoplasmic Ca(2+)/Mg(2+)-binding protein parvalbumin was highly abundant. NCX, PMCA, and CB prevailed in DCT2 and CNT, where we located the apical epithelial Ca(2+) channel (ECaC1). Its subcellular localization changed from apical in DCT2 to exclusively cytoplasmic at the end of CNT. NCX and PMCA decreased in parallel with the fading of ECaC1 in the apical membrane. All three of them were undetectable in CD. These findings disclose DCT2 and CNT as major sites for transcellular Ca(2+) transport in the mouse distal nephron. Cellular colocalization of Ca(2+) and Na(+) transport pathways suggests their mutual interactions in transport regulation.


Subject(s)
Calcium/metabolism , Carrier Proteins/analysis , Kidney Tubules, Distal/metabolism , Receptors, Drug , Sodium/metabolism , Symporters , Animals , Calbindins , Calcium Channels/analysis , Calcium Channels/immunology , Calcium-Transporting ATPases/analysis , Calcium-Transporting ATPases/immunology , Carrier Proteins/immunology , Cation Transport Proteins , Epithelial Sodium Channels , Female , Immunohistochemistry , Ion Transport , Kidney Tubules, Distal/chemistry , Mice , Models, Biological , Parvalbumins/analysis , Parvalbumins/immunology , Plasma Membrane Calcium-Transporting ATPases , S100 Calcium Binding Protein G/analysis , S100 Calcium Binding Protein G/immunology , Sodium Channels/analysis , Sodium Channels/immunology , Sodium Chloride Symporters , Sodium-Calcium Exchanger/analysis , Sodium-Calcium Exchanger/immunology , Solute Carrier Family 12, Member 3 , TRPV Cation Channels
18.
J Biol Chem ; 276(51): 47767-70, 2001 Dec 21.
Article in English | MEDLINE | ID: mdl-11687570

ABSTRACT

The calcium release-activated calcium channel (CRAC) is a highly Ca(2+)-selective ion channel that is activated on depletion of inositol triphosphate (IP(3))-sensitive intracellular Ca(2+) stores. It was recently reported that CaT1, a member of the TRP family of cation channels, exhibits the unique biophysical properties of CRAC, which led to the conclusion that CaT1 comprises all or part of the CRAC pore (Yue, L., Peng, J. B., Hediger, M. A., and Clapham, D. E. (2001) Nature 410, 705-709). Here, we directly compare endogenous CRAC with heterologously expressed CaT1 and show that they manifest several clearly distinct properties. CaT1 can be distinguished from CRAC in the following features: sensitivity to store-depleting agents; inward rectification in the absence of divalent cations; relative permeability to Na(+) and Cs(+); effect of 2-aminoethoxydiphenyl borate (2-APB). Moreover, CaT1 displays a mode of voltage-dependent gating that is fully absent in CRAC and originates from the voltage-dependent binding/unbinding of Mg(2+) inside the channel pore. Our results imply that the pores of CaT1 and CRAC are not identical and indicate that CaT1 is a Mg(2+)-gated channel not directly related to CRAC.


Subject(s)
Calcium Channels/physiology , Cell Line , Humans , Patch-Clamp Techniques , TRPV Cation Channels
19.
Proc Natl Acad Sci U S A ; 98(23): 13324-9, 2001 Nov 06.
Article in English | MEDLINE | ID: mdl-11687634

ABSTRACT

Rickets and hyperparathyroidism caused by a defective vitamin D receptor (VDR) can be prevented in humans and animals by high calcium intake, suggesting that intestinal calcium absorption is critical for 1,25(OH)(2) vitamin D [1,25(OH)(2)D(3)] action on calcium homeostasis. We assessed the rate of serum (45)Ca accumulation within 10 min of oral gavage in two strains of VDR-knockout (KO) mice (Leuven and Tokyo KO) and observed a 3-fold lower area under the curve in both KO strains. Moreover, we evaluated the expression of intestinal candidate genes involved in transcellular calcium transport. The calcium transport protein1 (CaT1) was more abundantly expressed at mRNA level than the epithelial calcium channel (ECaC) in duodenum, but both were considerably reduced (CaT1>90%, ECaC>60%) in the two VDR-KO strains on a normal calcium diet. Calbindin-D(9K) expression was decreased only in the Tokyo KO, whereas plasma membrane calcium ATPase (PMCA(1b)) expression was normal in both VDR-KOs. In Leuven wild-type mice, a high calcium diet inhibited (>90%) and 1,25(OH)(2)D(3) injection or low calcium diet induced (6-fold) duodenal CaT1 expression and, to a lesser degree, ECaC and calbindin-D(9K) expression. In Leuven KO mice, however, high or low calcium intake decreased calbindin-D(9K) and PMCA(1b) expression, whereas CaT1 and ECaC expression remained consistently low on any diet. These results suggest that the expression of the novel duodenal epithelial calcium channels (in particular CaT1) is strongly vitamin D-dependent, and that calcium influx, probably interacting with calbindin-D(9K), should be considered as a rate-limiting step in the process of vitamin D-dependent active calcium absorption.


Subject(s)
Calcium/metabolism , Duodenum/metabolism , Intestinal Absorption/genetics , Receptors, Calcitriol/physiology , Animals , Calcitriol/administration & dosage , Gene Expression , Kidney/metabolism , Mice , Mice, Knockout , Phenotype , Receptors, Calcitriol/genetics , Reverse Transcriptase Polymerase Chain Reaction
20.
Br J Pharmacol ; 134(3): 453-62, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11588099

ABSTRACT

1. The recent identification of the epithelial Ca(2+) channel, ECaC1, represents a major step forward in our knowledge of renal Ca(2+) handling. ECaC1 constitutes the rate-limiting apical Ca(2+) entry mechanism of active, transcellular Ca(2+) reabsorption. This unique highly selective Ca(2+) channel shares a low but significant homology with transient receptor potential (TRP) channels and vanilloid receptors (VR). 2. We have studied the pharmacological modulation of currents through ECaC1 heterologously expressed in HEK 293 cells. Monovalent cation currents were measured by use of the whole cell patch clamp technique in cells dialysed with 10 mM BAPTA or 10 mM EGTA to prevent the fast Ca(2+) dependent inactivation of ECaC1. 3. Several modulators were tested, including inorganic cations, putative store-operated Ca(2+) entry (SOC) blockers, the vanilloid receptor (VR-1) blocker capsazepine, protein tyrosine kinase blockers, calmodulin antagonists and ruthenium red. 4. Ruthenium red and econazole appeared to be the most effective inhibitors of currents through ECaC1, with IC(50) values of 111 nM and 1.3 microM, respectively, whereas the selective SOC inhibitor, SKF96365, was nearly ineffective. 5. The divalent cation current block profile for ECaC1 is Pb(2+)=Cu(2+) >Zn(2+) >Co(2+) >Fe(2+) with IC(50) values between 1 and approximately 10 microM. 6. In conclusion, ECaC activity is effectively inhibited by various compounds including ruthenium red, antimycotic drugs and divalent cations, which might be useful tools for pharmacological manipulation and several disorders related to Ca(2+) homeostasis could benefit from such developments.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Antifungal Agents/pharmacology , Cations, Monovalent/antagonists & inhibitors , Cations, Monovalent/metabolism , Cell Line/drug effects , Cell Line/physiology , Coloring Agents/pharmacology , Econazole/pharmacology , Epithelium/drug effects , Epithelium/metabolism , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Ruthenium Red/pharmacology , TRPV Cation Channels
SELECTION OF CITATIONS
SEARCH DETAIL
...