Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Trends Cell Biol ; 34(4): 299-311, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37865608

ABSTRACT

The rising prevalence of kidney diseases urges the need for novel therapies. Kidney organoids and tubuloids are advanced in vitro models and have recently been described as promising tools to study kidney (patho)physiology. Recent developments have shown their application in disease modeling, drug screening, and nephrotoxicity. These applications rely on their ability to mimic (dys)function in vitro including endocrine activity and drug, electrolyte, and water transport. This review provides an overview of these emerging kidney models and focuses on the most recent developments that utilize their functional capabilities. In addition, we cover current limitations and provide future perspectives for this rapidly evolving field, including what these functional properties mean for translational and personalized medicine now and in the future.


Subject(s)
Kidney , Organoids , Humans
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.
Chembiochem ; 15(10): 1446-51, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24904006

ABSTRACT

The ability of cells to incorporate azidosugars metabolically is a useful tool for extracellular glycan labelling. The exposed azide moiety can covalently react with alkynes, such as bicyclo[6.1.0]nonyne (BCN), by strain-promoted alkyne-azide cycloaddition (SPAAC). However, the use of SPAAC can be hampered by low specificity of the cycloalkyne. In this article we describe the synthesis of more polar BCN derivatives and their properties for selective cellular glycan labelling. The new polar derivatives [amino-BCN, glutarylamino-BCN and bis(hydroxymethyl)-BCN] display reaction rates similar to those of BCN and are less cell-permeable. The labelling specificity in HEK293 cells is greater than that of BCN, as determined by confocal microscopy and flow cytometry. Interestingly, amino-BCN appears to be highly specific for the Golgi apparatus. In addition, the polar BCN derivatives label the N-glycan of the membrane calcium channel TRPV5 in HEK293 cells with significantly enhanced signal-to-noise ratios.


Subject(s)
Alkynes/chemistry , Azides/chemistry , Bridged Bicyclo Compounds/chemical synthesis , Fluorescent Dyes/chemical synthesis , Polysaccharides/analysis , Bridged Bicyclo Compounds/analysis , Click Chemistry , Cycloaddition Reaction , Flow Cytometry , Fluorescent Dyes/analysis , Glycosylation , HEK293 Cells , Humans , Microscopy, Confocal , Optical Imaging , Polysaccharides/chemistry
5.
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
6.
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
7.
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
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.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...