Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
Sci Rep ; 12(1): 18551, 2022 11 03.
Article En | MEDLINE | ID: mdl-36329098

Digestion of dietary fibers by gut bacteria has been shown to stimulate intestinal mineral absorption [e.g., calcium (Ca2+) and magnesium (Mg2+)]. Although it has been suggested that local pH and short-chain fatty acid (SCFA) concentrations determine divalent cation absorption, the exact molecular mechanisms are still unknown. Therefore, this study aimed to determine the effects of SCFAs on intestinal Mg2+ absorption. We show that the butyrate concentration in the colon negatively correlates with serum Mg2+ levels in wildtype mice. Moreover, Na-butyrate significantly inhibited Mg2+ uptake in Caco-2 cells, while Ca2+ uptake was unaffected. Although Na-butyrate significantly lowered total ATP production rate, and resulted in increased phosphorylation of AMP-activated protein kinase (AMPK), inhibition of Mg2+ uptake by butyrate preceded these consequences. Importantly, electrophysiological examinations demonstrated that intracellular butyrate directly reduced the activity of the heteromeric Mg2+ channel complex, transient receptor potential melastatin (TRPM)6/7. Blocking cellular butyrate uptake prevented its inhibitory effect on Mg2+ uptake, demonstrating that butyrate acts intracellularly. Our work identified butyrate as novel regulator of intestinal Mg2+ uptake that works independently from metabolic regulation. This finding further highlights the role of microbial fermentation in the regulation of mineral absorption.


Butyrates , Magnesium , Humans , Mice , Animals , Butyrates/pharmacology , Butyrates/metabolism , Caco-2 Cells , Magnesium/metabolism , Colon/metabolism , Fatty Acids, Volatile/metabolism
2.
Acta Physiol (Oxf) ; 235(4): e13846, 2022 08.
Article En | MEDLINE | ID: mdl-35652564

Proton pump inhibitors (PPIs) reliably suppress gastric acid secretion and are therefore the first-line treatment for gastric acid-related disorders. Hypomagnesemia (serum magnesium [Mg2+ ] <0.7 mmol/L) is a commonly reported side effect of PPIs. Clinical reports demonstrate that urinary Mg2+ excretion is low in PPI users with hypomagnesemia, suggesting a compensatory mechanism by the kidney for malabsorption of Mg2+ in the intestines. However, the exact mechanism by which PPIs cause impaired Mg2+ absorption is still unknown. In this review, we show that current experimental evidence points toward reduced Mg2+ solubility in the intestinal lumen. Moreover, the absorption pathways in both the small intestine and the colon may be reduced by changes in the expression and activity of key transporter proteins. Additionally, the gut microbiome may contribute to the development of PPI-induced hypomagnesemia, as PPI use affects the composition of the gut microbiome. In this review, we argue that the increase of the luminal pH during PPI treatment may contribute to several of these mechanisms. Considering the fact that bacterial fermentation of dietary fibers results in luminal acidification, we propose that targeting the gut microbiome using dietary intervention might be a promising treatment strategy to restore hypomagnesemia in PPI users.


Gastrointestinal Microbiome , Proton Pump Inhibitors , Colon/metabolism , Homeostasis , Magnesium/metabolism , Magnesium/pharmacology , Proton Pump Inhibitors/adverse effects
3.
Pflugers Arch ; 474(3): 293-302, 2022 03.
Article En | MEDLINE | ID: mdl-34997297

Dietary fibers have been shown to increase the intestinal absorption of calcium (Ca2+) and magnesium (Mg2+). However, the mechanisms that explain the enhanced electrolyte absorption remain unknown. Therefore, this study aims to investigate the short-term and long-term effects of 5% (w/w) sodium butyrate (Na-butyrate), an important end-metabolite of bacterial fermentation of dietary fibers, on Ca2+ and Mg2+ homeostasis in mice. Serum Ca2+ levels were only significantly increased in mice treated with Na-butyrate for 1 day. This was associated with a twofold increase in the mRNA expression levels of Trpv6 in the proximal and distal colon. Contrary, Na-butyrate did not affect serum Mg2+ concentrations at either of the intervention periods. However, we observed a reduction in urinary Mg2+ excretion, although not significantly, after 1 day of treatment. A significant reduction of 2.5-fold in urinary Mg2+ excretion was observed after 14 days of treatment. Indeed, 14-day Na-butyrate supplementation increased colonic Trpm7 expression by 1.2-fold compared to control mice. In conclusion, short-term Na-butyrate supplementation increases serum Ca2+ levels in mice. This was associated with increased mRNA expression levels of Trpv6 in the colon, suggesting that Na-butyrate regulates the expression of genes involved in active intestinal Ca2+ absorption.


Sodium, Dietary , TRPM Cation Channels , Animals , Butyric Acid/pharmacology , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Colon , Dietary Fiber/metabolism , Dietary Fiber/pharmacology , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sodium/metabolism , Sodium Chloride, Dietary/metabolism , Sodium, Dietary/metabolism , Sodium, Dietary/pharmacology , TRPM Cation Channels/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
4.
Acta Biomater ; 99: 110-120, 2019 11.
Article En | MEDLINE | ID: mdl-31465881

Intestinal enterocytes are key players in the absorption of magnesium (Mg2+) and calcium (Ca2+). Understanding the exact molecular mechanisms by which their absorption behavior is regulated could greatly improve treatment strategies for stimulating intestinal absorption in diseases with Mg2+ and/or Ca2+ deficiency. However, such studies are hampered by the lack of in vitro intestinal cell models mimicking the mechanical and physiological properties of the gut. In this study we develop an in vitro gut model based on porous micropatterned membranes with villi-like surface topography and mechanical properties closely mimicking that of intestinal tissue. These membranes are prepared via phase separation micromolding using poly-ε-caprolactone/poly-lactic-glycolic acid (PCL/PLGA) polymer blend and can facilitate cellular differentiation of Caco-2 cells similar to native enterocytes. In fact, cells cultured on these micropatterned membranes form a brush border of microvilli with spatial differences in morphology and tight junction formation along the villous-base axis. Moreover, cells cultured on our membranes show a 2-fold increased alkaline phosphatase activity at the end of differentiation. Finally, we demonstrate that cells cultured on our micropatterned membranes have a 4- and 1.5-fold increased uptake of 25Mg and 45Ca, respectively, compared to non-patterned membranes. These results indicate that the new membranes can mimic the intestinal environment and therefore can have a great impact on mineral uptake in vitro. STATEMENT OF SIGNIFICANCE: This study presents the development of an in vitro gut model consisting of villi-like PCL/PLGA micropatterned membranes. These membranes are prepared via phase separation micromolding (PSµM), a technique which allows tailoring of the membrane surface topography combined with membrane porosity and interconnectivity which are important parameters for membranes used for in vitro transport studies. The culture of Caco-2 cells on these micropatterned membranes shows that they facilitate cellular differentiation similar to gut enterocytes. Our data indicate that mimicking the 3D geometry of the gut is very important for improving the physiological relevance of in vitro gut models. In the future, our micropatterned membranes with segment-specific geometries, in combination with isotopic measurements, would be applied to perform detailed ion uptake and transport studies.


Calcium/metabolism , Intestinal Mucosa/metabolism , Magnesium/metabolism , Tissue Scaffolds , Alkaline Phosphatase/metabolism , Biocompatible Materials , Caco-2 Cells , Cell Differentiation , Cell Proliferation , Enterocytes/metabolism , Humans , Microvilli/metabolism , Permeability , Polyesters/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Porosity , Surface Properties , Tight Junctions , Tissue Engineering/methods
5.
FASEB J ; 33(10): 11235-11246, 2019 10.
Article En | MEDLINE | ID: mdl-31299175

Proton pump inhibitors (PPIs) are used by millions of patients for the treatment of stomach acid-reflux diseases. Although PPIs are generally considered safe, about 13% of the users develop hypomagnesemia. Despite rising attention for this issue, the underlying mechanism is still unknown. Here, we examine whether the gut microbiome is involved in the development of PPI-induced hypomagnesemia in wild-type C57BL/6J mice. After 4 wk of treatment under normal or low dietary Mg2+ availability, omeprazole significantly reduced serum Mg2+ levels only in mice on a low-Mg2+ diet without affecting the mRNA expression of colonic or renal Mg2+ transporters. Overall, 16S rRNA gene sequencing revealed a lower gut microbial diversity in omeprazole-treated mice. Omeprazole induced a shift in microbial composition, which was associated with a 3- and 2-fold increase in the abundance of Lactobacillus and Bifidobacterium, respectively. To examine the metabolic consequences of these microbial alterations, the colonic composition of organic acids was evaluated. Low dietary Mg2+ intake, independent of omeprazole treatment, resulted in a 10-fold increase in formate levels. Together, these results imply that both omeprazole treatment and low dietary Mg2+ intake disturb the gut internal milieu and may pose a risk for the malabsorption of Mg2+ in the colon.-Gommers, L. M. M., Ederveen, T. H. A., van der Wijst, J., Overmars-Bos, C., Kortman, G. A. M., Boekhorst, J., Bindels, R. J. M., de Baaij, J. H. F., Hoenderop, J. G. J. Low gut microbiota diversity and dietary magnesium intake are associated with the development of PPI-induced hypomagnesemia.


Gastrointestinal Microbiome/physiology , Magnesium/metabolism , Proton Pump Inhibitors/adverse effects , Animals , Bifidobacterium/physiology , Colon/drug effects , Colon/metabolism , Colon/microbiology , Diet , Lactobacillus/physiology , Male , Mice , Mice, Inbred C57BL , Omeprazole/adverse effects , RNA, Ribosomal, 16S/metabolism
6.
PLoS One ; 14(6): e0217925, 2019.
Article En | MEDLINE | ID: mdl-31163064

There is an increasing amount of clinical evidence that hypomagnesemia (serum Mg2+ levels < 0.7 mmol/l) contributes to type 2 diabetes mellitus pathogenesis. Amongst other hypotheses, it has been suggested that Mg2+ deficiency affects insulin secretion. The aim of this study was, therefore, to investigate the acute effects of extracellular Mg2+ on glucose-stimulated insulin secretion in primary mouse islets of Langerhans and the rat insulinoma INS-1 cell line. Here we show that acute lowering of extracellular Mg2+ concentrations from 1.0 mM to 0.5 mM did not affect glucose-stimulated insulin secretion in islets or in insulin-secreting INS-1 cells. The expression of key genes in the insulin secretory pathway (e.g. Gck, Abcc8) was also unchanged in both experimental models. Knockdown of the most abundant Mg2+ channel Trpm7 by siRNAs in INS-1 cells resulted in a 3-fold increase in insulin secretion at stimulatory glucose conditions compared to mock-transfected cells. Our data suggest that insulin secretion is not affected by acute lowering of extracellular Mg2+ concentrations.


Extracellular Space/chemistry , Glucose/pharmacology , Insulin Secretion/drug effects , Magnesium/pharmacology , Animals , Gene Expression Regulation/drug effects , Hyperglycemia/pathology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , TRPM Cation Channels/metabolism
7.
Diabetes ; 65(1): 3-13, 2016 Jan.
Article En | MEDLINE | ID: mdl-26696633

Over the past decades, hypomagnesemia (serum Mg(2+) <0.7 mmol/L) has been strongly associated with type 2 diabetes mellitus (T2DM). Patients with hypomagnesemia show a more rapid disease progression and have an increased risk for diabetes complications. Clinical studies demonstrate that T2DM patients with hypomagnesemia have reduced pancreatic ß-cell activity and are more insulin resistant. Moreover, dietary Mg(2+) supplementation for patients with T2DM improves glucose metabolism and insulin sensitivity. Intracellular Mg(2+) regulates glucokinase, KATP channels, and L-type Ca(2+) channels in pancreatic ß-cells, preceding insulin secretion. Moreover, insulin receptor autophosphorylation is dependent on intracellular Mg(2+) concentrations, making Mg(2+) a direct factor in the development of insulin resistance. Conversely, insulin is an important regulator of Mg(2+) homeostasis. In the kidney, insulin activates the renal Mg(2+) channel transient receptor potential melastatin type 6 that determines the final urinary Mg(2+) excretion. Consequently, patients with T2DM and hypomagnesemia enter a vicious circle in which hypomagnesemia causes insulin resistance and insulin resistance reduces serum Mg(2+) concentrations. This Perspective provides a systematic overview of the molecular mechanisms underlying the effects of Mg(2+) on insulin secretion and insulin signaling. In addition to providing a review of current knowledge, we provide novel directions for future research and identify previously neglected contributors to hypomagnesemia in T2DM.


Diabetes Mellitus, Type 2/metabolism , Magnesium Deficiency/metabolism , Magnesium/metabolism , Water-Electrolyte Imbalance/metabolism , Blood Glucose/metabolism , Calcium Channels, L-Type/metabolism , Diabetes Mellitus, Type 2/complications , Dietary Supplements , Disease Progression , Glucokinase/metabolism , Glycogen/biosynthesis , Glycolysis , Humans , Inflammation , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Insulin-Secreting Cells/metabolism , KATP Channels/metabolism , Liver/metabolism , Magnesium/therapeutic use , Magnesium Deficiency/drug therapy , Obesity/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Sodium Chloride Symporters/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Water-Electrolyte Imbalance/drug therapy
8.
PLoS One ; 10(9): e0138881, 2015.
Article En | MEDLINE | ID: mdl-26397986

BACKGROUND: Proton-pump inhibitor-induced hypomagnesemia (PPIH) is the most recognized side effect of proton-pump inhibitors (PPIs). Additionally, PPIH is associated with hypocalcemia and hypokalemia. It is hypothesized that PPIs reduce epithelial proton secretion and thereby increase the pH in the colon, which may explain the reduced absorption of and Mg2+ and Ca2+. Fermentation of dietary oligofructose-enriched inulin fibers by the microflora leads to acidification of the intestinal lumen and by this enhances mineral uptake. This study aimed, therefore, to improve mineral absorption by application of dietary inulin to counteract PPIH. METHODS: Here, C57BL/J6 mice were supplemented with omeprazole and/or inulin. Subsequently, Mg2+ and Ca2+ homeostasis was assessed by means of serum, urine and fecal electrolyte measurements. Moreover, the mRNA levels of magnesiotropic and calciotropic genes were examined in the large intestine and kidney by real-time PCR. RESULTS: Treatment with omeprazole significantly reduced serum Mg2+ and Ca2+ levels. However, concomitant addition of dietary inulin fibers normalized serum Ca2+ but not serum Mg2+ concentrations. Inulin abolished enhanced expression of Trpv6 and S100g in the colon by omeprazole. Additionally, intestinal and renal mRNA levels of the Trpm6 gene were reduced after inulin intake. CONCLUSIONS: This study suggests that dietary inulin counteracts reduced intestinal Ca2+ absorption upon PPI treatment. In contrast, inulin did not increase intestinal absorption of Mg2+ sufficiently to recover serum Mg2+. The clinical potential of dietary inulin treatment should be the subject of future studies.


Dietary Fiber/administration & dosage , Hypocalcemia/prevention & control , Inulin/administration & dosage , Omeprazole/adverse effects , Proton Pump Inhibitors/adverse effects , Animals , Calcium/blood , Drug Evaluation, Preclinical , Fatty Acids/biosynthesis , Hypocalcemia/blood , Hypocalcemia/chemically induced , Intestinal Absorption/drug effects , Magnesium/blood , Male , Mice, Inbred C57BL , S100 Calcium Binding Protein G/metabolism
...