Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Eur J Nutr ; 58(2): 743-753, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29594477

RESUMEN

PURPOSE: There is general agreement that some dietary polyphenols block non-haem iron uptake, but the mechanisms by which they achieve this action are poorly understood. Since the polyphenol quercetin is ingested daily in significant amounts, we have investigated the effect of quercetin on duodenal non-haem iron absorption in vivo, as well as its effect on factors known to be involved in systemic iron metabolism. METHODS: Rats were subject to gastric gavage and systemic quercetin administration. Treatments were followed with uptake studies using radiolabeled iron, serum iron and transferrin saturation measurements, LC-MS/MS analysis of quercetin metabolites in serum, determination of tissue non-haem iron content and analysis of gene expression of iron-related proteins. RESULTS: Both oral and intraperitoneal (IP) quercetin caused serum and tissue iron depletion by two means, first by increasing mucosal iron uptake and inhibiting iron efflux from duodenal mucosa, and second by decreasing levels of duodenal DMT1, Dcytb and FPN. Additionally, IP quercetin induced highly significant increased liver expression of hepcidin, a hormone known to inhibit intestinal iron uptake. CONCLUSIONS: Oral quercetin significantly inhibited iron absorption, while IP quercetin significantly affected iron-related genes. These results could lead to development of new effective ways of preventing and treating iron deficiency anaemia, the most widespread nutritional disorder in the world.


Asunto(s)
Antioxidantes/farmacología , Duodeno/efectos de los fármacos , Duodeno/metabolismo , Expresión Génica/efectos de los fármacos , Hierro/metabolismo , Quercetina/farmacología , Animales , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Masculino , Modelos Animales , Ratas , Ratas Sprague-Dawley
2.
Exp Physiol ; 102(4): 462-474, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28130860

RESUMEN

NEW FINDINGS: What is the central question of this study? Does a previously hypothesized signalling mechanism, believed to detect postprandial increases in intestinal phosphate and that can stimulate the kidneys to rapidly excrete phosphate, operate under physiological conditions? What is the main finding and its importance? Contrary to earlier reports, rapid signalling between the small intestine and kidney mediated by a gut-derived phosphaturic factor in response to a physiological intestinal phosphate load is not supported by the present findings; moreover, hyperphosphataemia and increased parathyroid hormone concentrations are likely to be the underlying factors responsible for the phosphaturia following a supraphysiological intestinal phosphate load. To date, the role of the small intestine in the regulation of postprandial phosphate homeostasis has remained unclear and controversial. Previous studies have proposed the presence of a gut-derived phosphaturic factor that acts independently of changes in plasma phosphate concentration or parathyroid hormone (PTH) concentration; however, these early studies used duodenal luminal phosphate concentrations in the molar range, and therefore, the physiological relevance of this is uncertain. In the present study, we used both in vivo and in vitro approaches to investigate the presence of this putative 'intestinal phosphatonin'. Instillation of 1.3 m phosphate into the duodenum rapidly induced phosphaturia, but in contrast to previous reports, this was associated with significant hyperphosphataemia and elevated PTH concentration; however, there was not the expected decrease in abundance of the renal sodium-phosphate cotransporter NaPi-IIa. Instillation of a physiological (10 mm) phosphate load had no effect on plasma phosphate concentration, PTH concentration or phosphate excretion. Moreover, phosphate uptake by opossum kidney cells was unaffected after incubation with serosal fluid collected from intestinal segments perfused with different concentrations of phosphate. Taken together, these findings do not support the concept of a gut-derived phosphaturic factor that can mediate rapid signalling between the gut and kidney, leading to increased urinary phosphate excretion, as part of normal phosphate homeostasis.


Asunto(s)
Mucosa Intestinal/metabolismo , Riñón/metabolismo , Riñón/fisiología , Fosfatos/metabolismo , Periodo Posprandial/fisiología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/metabolismo , Animales , Transporte Biológico/fisiología , Homeostasis/fisiología , Masculino , Hormona Paratiroidea/metabolismo , Ratas , Ratas Sprague-Dawley , Simportadores/metabolismo
3.
Biochem Biophys Res Commun ; 469(3): 521-8, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26655811

RESUMEN

The liver expresses batteries of cytoprotective genes that confer cellular resistance to oxidative stress and xenobiotic toxins, and protection against cancer and other stress-related diseases. These genes are mainly regulated by Nrf2, making this transcription factor a target for small molecule discovery to treat such diseases. In this report, we identified dietary polyphenolic antioxidants that not only activated these genes but also relieved Nrf2 repression by Keap1, a Cul3-dependent ubiquitin ligase adaptor protein that mediates its degradation. Analysis of postprandial liver RNA revealed a marked activation of both genes by all test polyphenols compared with controls. Nrf2 inhibition by RNA interference reduced polyphenol effects on its target gene expression. Our data suggest that polyphenols may induce cellular defense genes by derepressing Nrf2 inhibition by Keap1. We posit that this ability to derepress Nrf2 and reactivate its target genes may underlie the protection conferred by polyphenols against oxidative stress-related diseases.


Asunto(s)
Hepatocitos/efectos de los fármacos , Hepatocitos/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Polifenoles/administración & dosificación , Transcripción Genética/fisiología , Administración Oral , Animales , Suplementos Dietéticos , Regulación de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína 1 Asociada A ECH Tipo Kelch , Masculino , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transcripción Genética/efectos de los fármacos
4.
Exp Physiol ; 101(6): 731-42, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27164183

RESUMEN

What is the central question of this study? Although SGLT2 inhibitors represent a promising treatment for patients suffering from diabetic nephropathy, the influence of metabolic disruption on the expression and function of glucose transporters is largely unknown. What is the main finding and its importance? In vivo models of metabolic disruption (Goto-Kakizaki type II diabetic rat and junk-food diet) demonstrate increased expression of SGLT1, SGLT2 and GLUT2 in the proximal tubule brush border. In the type II diabetic model, this is accompanied by increased SGLT- and GLUT-mediated glucose uptake. A fasted model of metabolic disruption (high-fat diet) demonstrated increased GLUT2 expression only. The differential alterations of glucose transporters in response to varying metabolic stress offer insight into the therapeutic value of inhibitors. SGLT2 inhibitors are now in clinical use to reduce hyperglycaemia in type II diabetes. However, renal glucose reabsorption across the brush border membrane (BBM) is not completely understood in diabetes. Increased consumption of a Western diet is strongly linked to type II diabetes. This study aimed to investigate the adaptations that occur in renal glucose transporters in response to experimental models of diet-induced insulin resistance. The study used Goto-Kakizaki type II diabetic rats and normal rats rendered insulin resistant using junk-food or high-fat diets. Levels of protein kinase C-ßI (PKC-ßI), GLUT2, SGLT1 and SGLT2 were determined by Western blotting of purified renal BBM. GLUT- and SGLT-mediated d-[(3) H]glucose uptake by BBM vesicles was measured in the presence and absence of the SGLT inhibitor phlorizin. GLUT- and SGLT-mediated glucose transport was elevated in type II diabetic rats, accompanied by increased expression of GLUT2, its upstream regulator PKC-ßI and SGLT1 protein. Junk-food and high-fat diet feeding also caused higher membrane expression of GLUT2 and its upstream regulator PKC-ßI. However, the junk-food diet also increased SGLT1 and SGLT2 levels at the proximal tubule BBM. Glucose reabsorption across the proximal tubule BBM, via GLUT2, SGLT1 and SGLT2, is not solely dependent on glycaemic status, but is also influenced by diet-induced changes in glucose metabolism. We conclude that different metabolic disturbances result in complex adaptations in renal glucose transporter protein levels and function.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Túbulos Renales Proximales/metabolismo , Membranas/metabolismo , Animales , Glucemia/metabolismo , Dieta Alta en Grasa/efectos adversos , Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/metabolismo , Hiperglucemia/metabolismo , Resistencia a la Insulina/fisiología , Riñón/metabolismo , Masculino , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley , Transportador 1 de Sodio-Glucosa/metabolismo , Transportador 2 de Sodio-Glucosa/metabolismo
5.
Curr Opin Nephrol Hypertens ; 22(4): 481-7, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23666413

RESUMEN

PURPOSE OF REVIEW: For a number of years, there has been increasing interest in the concept of directly targeting intestinal phosphate transport to control hyperphosphatemia in chronic kidney disease. However, progress has been slow due to the paucity of information on the mechanisms involved in intestinal phosphate absorption. This editorial highlights the most recent developments in our understanding of this process and the role of the intestine in the maintenance of phosphate balance. RECENT FINDINGS: Recent studies in NaPi-IIb knockout mice have confirmed that this transport protein plays a significant role in intestinal phosphate absorption and is critical in the proposed feed-forward mechanism between the small intestine and kidney, which helps to maintain normal phosphate balance and steady-state plasma phosphate concentrations. In addition, renal failure-induced hyperphosphatemia is attenuated in NaPi-IIb knockout mice, confirming that NaPi-IIb is a suitable target in the prevention and treatment of hyperphosphatemia. SUMMARY: Recent findings suggest that consumption of processed foods containing phosphate preservatives may lead to excessive phosphate exposure (if not overload), toxicity, and cardiovascular disease in the general population, as well as in patients with declining renal function. Therefore, establishing more effective ways of targeting the intestine to limit dietary phosphate absorption could have wide-reaching health benefits.


Asunto(s)
Conservantes de Alimentos/metabolismo , Hiperfosfatemia/metabolismo , Absorción Intestinal , Mucosa Intestinal/metabolismo , Fosfatos/metabolismo , Fósforo Dietético/metabolismo , Insuficiencia Renal Crónica/metabolismo , Animales , Conservantes de Alimentos/efectos adversos , Homeostasis , Humanos , Hiperfosfatemia/genética , Hiperfosfatemia/fisiopatología , Hiperfosfatemia/terapia , Intestinos/fisiopatología , Ratones , Ratones Noqueados , Fosfatos/efectos adversos , Fósforo Dietético/efectos adversos , Insuficiencia Renal Crónica/fisiopatología , Insuficiencia Renal Crónica/terapia , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIb/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIb/metabolismo
6.
Am J Physiol Endocrinol Metab ; 303(5): E669-81, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22811473

RESUMEN

The inhibitory effects of the angiotensin-converting enzyme (ACE)-ANG II-angiotensin type 1 (AT1) receptor axis on jejunal glucose uptake and the reduced expression of this system in type 1 diabetes mellitus (T1DM) have been documented previously. The ACE2-ANG-(1-7)-Mas receptor axis is thought to oppose the actions of the ACE-ANG II-AT1 receptor axis in heart, liver, and kidney. However, the possible involvement of the ACE2-ANG-(1-7)-Mas receptor system on enhanced jejunal glucose transport in T1DM has yet to be determined. Rat everted jejunum and Caco-2 cells were used to determine the effects of ANG-(1-7) on glucose uptake and to study the ACE2-ANG-(1-7)-Mas receptor signaling pathway. Expression of target gene and protein in jejunal enterocytes and human Caco-2 cells were quantified using real-time PCR and Western blotting. T1DM increased jejunal protein and mRNA expression of ACE2 (by 59 and 173%, respectively) and Mas receptor (by 55 and 100%, respectively) in jejunum. One millimolar ANG-(1-7) reduced glucose uptake in jejunum and Caco-2 cells by 30.6 and 30.3%, respectively, effects that were abolished following addition of 1 µM A-779 (a Mas receptor blocker) or 1 µM GF-109203X (protein kinase C inhibitor) to incubation buffer for jejunum or Caco-2 cells, respectively. Finally, intravenous treatment of animals with ANG-(1-7) significantly improved oral glucose tolerance in T1DM but not control animals. In conclusion, enhanced activity of the ACE2-ANG-(1-7)-Mas receptor axis in jejunal enterocytes is likely to moderate the T1DM-induced increase in jejunal glucose uptake resulting from downregulation of the ACE-ANG II-AT1 receptor axis. Therefore, altered activity of both ACE and ACE2 systems during diabetes will determine the overall rate of glucose transport across the jejunal epithelium.


Asunto(s)
Angiotensina I/metabolismo , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Enterocitos/metabolismo , Yeyuno/metabolismo , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Regulación hacia Arriba , Angiotensina I/administración & dosificación , Angiotensina I/genética , Angiotensina I/uso terapéutico , Enzima Convertidora de Angiotensina 2 , Animales , Transporte Biológico/efectos de los fármacos , Células CACO-2 , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Enterocitos/efectos de los fármacos , Enterocitos/patología , Glucosa/metabolismo , Humanos , Hiperglucemia/prevención & control , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/metabolismo , Hipoglucemiantes/uso terapéutico , Técnicas In Vitro , Inyecciones Intravenosas , Yeyuno/efectos de los fármacos , Yeyuno/patología , Masculino , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/uso terapéutico , Peptidil-Dipeptidasa A/genética , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética
7.
Br J Nutr ; 105(11): 1607-18, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21411025

RESUMEN

The secoiridoids 3,4-dihydroxyphenylethanol-elenolic acid (3,4-DHPEA-EA) and 3,4-dihydroxyphenylethanol-elenolic acid dialdehyde (3,4-DHPEA-EDA) account for approximately 55 % of the phenolic content of olive oil and may be partly responsible for its reported human health benefits. We have investigated the absorption and metabolism of these secoiridoids in the upper gastrointestinal tract. Both 3,4-DHPEA-EDA and 3,4-DHPEA-EA were relatively stable under gastric conditions, only undergoing limited hydrolysis. Both secoiridoids were transferred across a human cellular model of the small intestine (Caco-2 cells). However, no glucuronide conjugation was observed for either secoiridoid during transfer, although some hydroxytyrosol and homovanillic alcohol were formed. As Caco-2 cells are known to express only limited metabolic activity, we also investigated the absorption and metabolism of secoiridoids in isolated, perfused segments of the jejunum and ileum. Here, both secoiridoids underwent extensive metabolism, most notably a two-electron reduction and glucuronidation during the transfer across both the ileum and jejunum. Unlike Caco-2 cells, the intact small-intestinal segments contain NADPH-dependent aldo-keto reductases, which reduce the aldehyde carbonyl group of 3,4-DHPEA-EA and one of the two aldeydic carbonyl groups present on 3,4-DHPEA-EDA. These reduced forms are then glucuronidated and represent the major in vivo small-intestinal metabolites of the secoiridoids. In agreement with the cell studies, perfusion of the jejunum and ileum also yielded hydroxytyrosol and homovanillic alcohol and their respective glucuronides. We suggest that the reduced and glucuronidated forms represent novel physiological metabolites of the secoiridoids that should be pursued in vivo and investigated for their biological activity.


Asunto(s)
Intestino Delgado/metabolismo , Fenoles/metabolismo , Aceites de Plantas/química , Piranos/metabolismo , Animales , Transporte Biológico , Células CACO-2 , Humanos , Concentración de Iones de Hidrógeno , Absorción Intestinal , Modelos Biológicos , Estructura Molecular , Aceite de Oliva , Fenoles/química , Piranos/química , Ratas
8.
Am J Physiol Renal Physiol ; 299(2): F285-96, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20534868

RESUMEN

Transport of phosphate across intestinal and renal epithelia is essential for normal phosphate balance, yet we know less about the mechanisms and regulation of intestinal phosphate absorption than we do about phosphate handling by the kidney. Recent studies have provided strong evidence that the sodium-phosphate cotransporter NaPi-IIb is responsible for sodium-dependent phosphate absorption by the small intestine, and it might be that this protein can link changes in dietary phosphate to altered renal phosphate excretion to maintain phosphate balance. Evidence is also emerging that specific regions of the small intestine adapt differently to acute or chronic changes in dietary phosphate load and that phosphatonins inhibit both renal and intestinal phosphate transport. This review summarizes our current understanding of the mechanisms and control of intestinal phosphate absorption and how it may be related to renal phosphate reabsorption; it also considers the ways in which the gut could be targeted to prevent, or limit, hyperphosphatemia in chronic and end-stage renal failure.


Asunto(s)
Absorción Intestinal , Intestino Delgado/metabolismo , Riñón/metabolismo , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato/metabolismo , Animales , Células Epiteliales/metabolismo , Homeostasis , Humanos , Hiperfosfatemia/etiología , Hiperfosfatemia/metabolismo , Hiperfosfatemia/prevención & control , Mucosa Intestinal/metabolismo , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/metabolismo , Fosfatos/sangre
9.
Kidney Int ; 78(7): 660-7, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20631677

RESUMEN

Erythropoietin is produced by the kidney and stimulates erythropoiesis; however, in chronic renal disease its levels are reduced and patients develop anemia that is treatable with iron and recombinant hormone. The mechanism by which erythropoietin improves iron homeostasis is still unclear, but it may involve suppression of the iron regulatory peptide hepcidin and/or a direct effect on intestinal iron absorption. To investigate these possibilities, we used the well-established 5/6th nephrectomy rat model of chronic renal failure with or without human recombinant erythropoietin treatment. Monolayers of human intestinal Caco-2 cells were also treated with erythropoietin to measure any direct effects of this hormone on intestinal iron transport. Nephrectomy increased hepatic hepcidin expression and decreased intestinal iron absorption; these effects were restored to levels found in sham-operated rats on erythropoietin treatment of the rats with renal failure. In Caco-2 cells, the addition of erythropoietin significantly increased the expression of apical divalent metal transporter 1 (DMT1) and basolateral ferroportin and, consequently, iron transport across the monolayer. Taken together, our results show that erythropoietin not only exerts a powerful inhibitory action on the expression of hepcidin, thus permitting the release of iron from reticuloendothelial macrophages and intestinal enterocytes, but also acts directly on enterocytes to increase iron absorption.


Asunto(s)
Eritropoyetina/farmacología , Absorción Intestinal/efectos de los fármacos , Hierro/metabolismo , Fallo Renal Crónico/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/genética , Células CACO-2 , Proteínas de Transporte de Catión/genética , Modelos Animales de Enfermedad , Duodeno/metabolismo , Hepcidinas , Humanos , Masculino , Nefrectomía , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Receptores de Eritropoyetina/análisis , Transducción de Señal
10.
Am J Physiol Cell Physiol ; 297(3): C601-10, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19535516

RESUMEN

Streptozotocin-induced (Type 1) diabetes mellitus (T1DM) in rats promotes jejunal glucose transport, but the trigger for this response remains unclear. Our recent work using euglycemic rats has implicated the enterocyte renin-angiotensin system (RAS) in control of sodium-dependent glucose transporter (SGLT1)-mediated glucose uptake across the jejunal brush border membrane (BBM). The aim of the present study was to examine whether expression of enterocyte RAS components is influenced by T1DM. The effects of mucosal addition of angiotensin II (AII) on [(14)C]-D-glucose uptake by everted diabetic jejunum was also determined. Two-week diabetes caused a fivefold increase in blood glucose level and reduced mRNA and protein expression of AII type 1 (AT(1)) and AT(2) receptors and angiotensin-converting enzyme in isolated jejunal enterocytes. Angiotensinogen expression was, however, stimulated by diabetes while renin was not detected in either control or diabetic enterocytes. Diabetes stimulated glucose uptake into everted jejunum by 58% and increased the BBM expression of SGLT1 and facilitated glucose transporter 2 (GLUT2) proteins, determined by Western blotting by 25% and 135%, respectively. Immunohistochemistry confirmed an enhanced BBM expression of GLUT2 in diabetes and also showed that this was due to translocation of the transporter from the basolateral membrane to BBM. AII (5 microM) or L-162313 (1 microM), a nonpeptide AII analog, decreased glucose uptake by 18% and 24%, respectively, in diabetic jejunum. This inhibitory action was fully accountable by an action on SGLT1-mediated transport and was abolished by the AT(1) receptor antagonist losartan (1 microM). The decreased inhibitory action of AII on in vitro jejunal glucose uptake in diabetes compared with that noted previously in jejunum from normal animals is likely to be due to reduced RAS expression in diabetic enterocytes, together with a disproportionate increase in GLUT2, compared with SGLT1 expression at the BBM.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Enterocitos/metabolismo , Glucosa/metabolismo , Microvellosidades/fisiología , Sistema Renina-Angiotensina/fisiología , Animales , Transporte Biológico Activo , Glucemia/metabolismo , Regulación de la Expresión Génica/fisiología , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Yeyuno/citología , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Transportador 1 de Sodio-Glucosa/genética , Transportador 1 de Sodio-Glucosa/metabolismo
11.
J Am Soc Nephrol ; 19(12): 2313-20, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19005008

RESUMEN

The role of putative humoral factors, known as phosphatonins, in phosphate homeostasis and the relationship between phosphate handling by the kidney and gastrointestinal tract are incompletely understood. Matrix extracellular phosphoglycoprotein (MEPE), one of several candidate phosphatonins, promotes phosphaturia, but whether it also affects intestinal phosphate absorption is unknown. Here, using the in situ intestinal loop technique, we demonstrated that short-term infusion of MEPE inhibits phosphate absorption in the jejunum but not the duodenum. Simultaneous measurement of urinary phosphate excretion suggests that the phosphaturic action of MEPE correlates with a significant reduction in the protein levels of the renal sodium-phosphate co-transporter NaPi-IIa in the proximal convoluted tubules of the outer renal cortex, assessed by Western blotting and immunohistochemistry. This short-term inhibitory effect of MEPE on renal and intestinal phosphate handling occurred without any changes in circulating levels of parathyroid hormone, 1,25-dihydroxyvitamin D(3), or fibroblast growth factor 23. Taken together, these findings suggest that MEPE is a candidate phosphatonin involved in phosphate homeostasis, acting in both the kidney and the gastrointestinal tract.


Asunto(s)
Proteínas de la Matriz Extracelular/fisiología , Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Fosfatos/metabolismo , Fosfoproteínas/fisiología , Animales , Proteínas de la Matriz Extracelular/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Tracto Gastrointestinal/metabolismo , Glicoproteínas/fisiología , Inmunohistoquímica/métodos , Corteza Renal/metabolismo , Masculino , Hormona Paratiroidea/metabolismo , Fosfatos/química , Fosfoproteínas/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley
12.
Physiol Rep ; 6(14): e13804, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30009546

RESUMEN

Artificial sweeteners are extensively used by the food industry to replace sugar in food and beverages and are widely considered to be a healthy alternative. However, recent data suggest that artificial sweeteners may impact intestinal glucose absorption and that they might lead to glucose intolerance. Moreover, chronic consumption of artificial sweeteners has also been linked to detrimental changes in renal function. Using an in vivo approach, our study aimed to determine if short-term infusion of the artificial sweetener saccharin can alter renal function and renal glucose absorption. We show that saccharin infusion does not induce any major change in GFR or urine flow rate at either the whole kidney or single nephron level, suggesting that any reported change in renal function with artificial sweeteners must depend on chronic consumption. As expected for a nondiabetic animal, glucose excretion was low; however, saccharin infusion caused a small, but significant, decrease in fractional glucose excretion. In contrast to the whole kidney data, our micropuncture results did not show any significant difference in fractional glucose reabsorption in either the proximal or distal tubules, indicating that saccharin does not influence renal glucose handling in vivo under euglycemic conditions. In keeping with this finding, protein levels of the renal glucose transporters SGLT1 and SGLT2 were also unchanged. In addition, saccharin infusion in rats undergoing a glucose tolerance test failed to induce a robust change in renal glucose excretion or renal glucose transporter expression. In conclusion, our results demonstrate that saccharin does not induce acute physiologically relevant changes in renal function or renal glucose handling.


Asunto(s)
Glucosa/metabolismo , Riñón/efectos de los fármacos , Reabsorción Renal , Sacarina/farmacología , Edulcorantes/farmacología , Animales , Riñón/metabolismo , Masculino , Ratas , Ratas Wistar , Sacarina/administración & dosificación , Sacarina/efectos adversos , Transportador 1 de Sodio-Glucosa/metabolismo , Transportador 2 de Sodio-Glucosa/metabolismo , Edulcorantes/administración & dosificación , Edulcorantes/efectos adversos
13.
Free Radic Biol Med ; 40(2): 323-34, 2006 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16413414

RESUMEN

Studies have suggested that diets rich in polyphenols such as flavonoids may lead to a reduced risk of gastrointestinal cancers. We demonstrate the ability of monomeric and dimeric flavanols to scavenge reactive nitrogen species derived from nitrous acid. Both epicatechin and dimer B2 (epicatechin dimer) inhibited nitrous acid-induced formation of 3-nitrotyrosine and the formation of the carcinogenic N-nitrosamine, N-nitrosodimethylamine. The reaction of monomeric and dimeric epicatechin with nitrous acid led to the formation of mono- and di-nitroso flavanols, whereas the reaction with hesperetin resulted primarily in the formation of nitrated products. Although, epicatechin was transferred across the jejunum of the small intestine yielding metabolites, its nitroso form was not absorbed. Dimer B2 but not epicatechin monomer inhibited the proliferation of, and triggered apoptosis in, Caco-2 cells. The latter was accompanied by caspase-3 activation and reductions in Akt phosphorylation, suggesting activation of apoptosis via inhibition of prosurvival signaling. Furthermore, the dinitroso derivative of dimer B2, and to a lesser extent the dinitroso-epicatechin, also induced significant toxic effects in Caco-2 cells. The inhibitory effects on cellular proliferation were paralleled by early inhibition of ERK 1/2 phosphorylation and later reductions in cyclin D1 levels, indicating modulation of cell cycle regulation in Caco-2 cells. These effects highlight multiple routes in which dietary derived flavanols may exert beneficial effects in the gastrointestinal tract.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Flavonoides/química , Flavonoides/farmacología , Compuestos Nitrosos/metabolismo , Compuestos Nitrosos/farmacología , Ácido Nitroso/química , Absorción , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Células CACO-2 , Caspasa 3 , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Catequina/análogos & derivados , Catequina/química , Catequina/farmacología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Ciclina D1/efectos de los fármacos , Ciclina D1/metabolismo , Dimetilnitrosamina , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Tracto Gastrointestinal/efectos de los fármacos , Humanos , Técnicas In Vitro , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Nitrosaminas/antagonistas & inhibidores , Nitrosaminas/química , Nitrosaminas/metabolismo , Compuestos Nitrosos/química , Ácido Nitroso/antagonistas & inhibidores , Fenoles/química , Fenoles/farmacología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Especies de Nitrógeno Reactivo/antagonistas & inhibidores , Especies de Nitrógeno Reactivo/farmacología , Factores de Tiempo , Tirosina/análogos & derivados , Tirosina/antagonistas & inhibidores , Tirosina/metabolismo
14.
Free Radic Res ; 40(6): 647-58, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16753843

RESUMEN

We have conducted a detailed investigation into the absorption, metabolism and microflora-dependent transformation of hydroxytyrosol (HT), tyrosol (TYR) and their conjugated forms, such as oleuropein (OL). Conjugated forms underwent rapid hydrolysis under gastric conditions, resulting in significant increases in the amount of free HT and TYR entering the small intestine. Both HT and TYR transferred across human Caco-2 cell monolayers and rat segments of jejunum and ileum and were subject to classic phase I/II biotransformation. The major metabolites identified were an O-methylated derivative of HT, glucuronides of HT and TYR and a novel glutathionylated conjugate of HT. In contrast, there was no absorption of OL in either model. However, OL was rapidly degraded by the colonic microflora resulting in the formation of HT. Our study provides additional information regarding the breakdown of complex olive oil polyphenols in the GI tract, in particular the stomach and the large intestine.


Asunto(s)
Flavonoides/farmacología , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Fenoles/farmacología , Aceites de Plantas/química , Animales , Biotransformación , Células CACO-2 , Cromatografía Líquida de Alta Presión , Colon/efectos de los fármacos , Colon/metabolismo , Colon/microbiología , Fermentación , Flavonoides/química , Ácido Gástrico/metabolismo , Tracto Gastrointestinal/metabolismo , Humanos , Absorción Intestinal , Estructura Molecular , Aceite de Oliva , Perfusión , Fenoles/química , Polifenoles , Ratas
15.
Physiol Rep ; 3(1)2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25626876

RESUMEN

Despite the importance of extracellular phosphate in many essential biological processes, the mechanisms of phosphate transport across the epithelium of different intestinal segments remain unclear. We have used an in vitro method to investigate phosphate transport at the brush border membrane (BBM) of intact intestinal segments and an in vivo method to study transepithelial phosphate absorption. We have used micromolar phosphate concentrations known to favor NaPi-IIb-mediated transport, and millimolar concentrations that are representative of the levels we have measured in luminal contents, to compare the extent of Na(+)-dependent and Na(+)-independent phosphate transport along the rat duodenum, jejunum, ileum, and proximal and distal colon. Our findings confirm that overall the jejunum is the main site of phosphate absorption; however, at millimolar concentrations, absorption shows ~30% Na(+)-dependency, suggesting that transport is unlikely to be mediated exclusively by the Na(+)-dependent NaPi-IIb co-transporter. In the ileum, studies in vitro confirmed that relatively low levels of phosphate transport occur at the BBM of this segment, although significant Na(+)-dependent transport was detected using millimolar levels of phosphate in vivo. Since NaPi-IIb protein is not detectable at the rat ileal BBM, our data suggest the presence of an as yet unidentified Na(+)-dependent uptake pathway in this intestinal segment in vivo. In addition, we have confirmed that the colon has a significant capacity for phosphate absorption. Overall, this study highlights the complexities of intestinal phosphate absorption that can be revealed using different phosphate concentrations and experimental techniques.

16.
Free Radic Biol Med ; 36(2): 212-25, 2004 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-14744633

RESUMEN

The metabolism of chlorogenic acid, naringin, and rutin, representative members of three common families of dietary polyphenols, the hydroxycinnamates, the flavanones, and the flavonols, respectively, was studied in an in vitro mixed culture model of the human colonic microflora. Time- and concentration-dependent degradation of all three compounds was observed, which was associated with the following metabolic events after cleavage of the ester or glycosidic bond: reduction of the aliphatic double bond of the resulting hydroxycinnamate caffeic acid residue; dehydroxylation and ring fission of the heterocyclic C-ring of the resulting deglycosylated flavanone, naringenin, and of the deglycosylated flavonol, quercetin (which differed depending on the substitution). The metabolic events, their sequences, and major phenolic end products, as identified by GC-MS or LC-MS/MS, were elucidated from the structural characteristics of the investigated compounds. The major phenolic end products identified were 3-(3-hydroxyphenyl)-propionic acid for chlorogenic acid, 3-(4-hydroxyphenyl)-propionic acid and 3-phenylpropionic acid for naringin, and 3-hydroxyphenylacetic acid and 3-(3-hydroxyphenyl)-propionic acid for rutin. The degree of degradation of the compounds studied was significantly influenced by the substrate concentration as well as individual variations in the composition of the fecal flora. The results support extensive metabolism of dietary polyphenols in the colon, depending on substrate concentration and residence time, with resultant formation of simple phenolics, which can be considered biomarkers of colonic metabolism if subsequently absorbed. It is also apparent that a relatively small number of phenolic degradation products are formed in the colon from the diverse group of natural polyphenols.


Asunto(s)
Colon/metabolismo , Colon/microbiología , Dieta , Fermentación/fisiología , Flavonoides/química , Flavonoides/metabolismo , Fenoles/química , Fenoles/metabolismo , Cromatografía Líquida de Alta Presión , Ácidos Cumáricos/química , Ácidos Cumáricos/metabolismo , Heces/química , Heces/microbiología , Flavanonas/química , Flavanonas/metabolismo , Flavonoides/administración & dosificación , Flavonoles/química , Flavonoles/metabolismo , Humanos , Fenoles/administración & dosificación , Polifenoles , Factores de Tiempo
17.
PLoS One ; 9(7): e102900, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25058155

RESUMEN

Balancing systemic iron levels within narrow limits is critical for maintaining human health. There are no known pathways to eliminate excess iron from the body and therefore iron homeostasis is maintained by modifying dietary absorption so that it matches daily obligatory losses. Several dietary factors can modify iron absorption. Polyphenols are plentiful in human diet and many compounds, including quercetin--the most abundant dietary polyphenol--are potent iron chelators. The aim of this study was to investigate the acute and longer-term effects of quercetin on intestinal iron metabolism. Acute exposure of rat duodenal mucosa to quercetin increased apical iron uptake but decreased subsequent basolateral iron efflux into the circulation. Quercetin binds iron between its 3-hydroxyl and 4-carbonyl groups and methylation of the 3-hydroxyl group negated both the increase in apical uptake and the inhibition of basolateral iron release, suggesting that the acute effects of quercetin on iron transport were due to iron chelation. In longer-term studies, rats were administered quercetin by a single gavage and iron transporter expression measured 18 h later. Duodenal FPN expression was decreased in quercetin-treated rats. This effect was recapitulated in Caco-2 cells exposed to quercetin for 18 h. Reporter assays in Caco-2 cells indicated that repression of FPN by quercetin was not a transcriptional event but might be mediated by miRNA interaction with the FPN 3'UTR. Our study highlights a novel mechanism for the regulation of iron bioavailability by dietary polyphenols. Potentially, diets rich in polyphenols might be beneficial for patients groups at risk of iron loading by limiting the rate of intestinal iron absorption.


Asunto(s)
Proteínas de Transporte de Catión/antagonistas & inhibidores , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Quelantes del Hierro/farmacología , Hierro de la Dieta/metabolismo , Quercetina/farmacología , Regiones no Traducidas 3' , Animales , Células CACO-2 , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Duodeno/efectos de los fármacos , Duodeno/metabolismo , Expresión Génica/efectos de los fármacos , Homeostasis/fisiología , Humanos , Absorción Intestinal/fisiología , Mucosa Intestinal/metabolismo , Transporte Iónico/efectos de los fármacos , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Ratas , Ratas Sprague-Dawley
18.
J Physiol ; 584(Pt 2): 613-23, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17702818

RESUMEN

There is increasing evidence that locally produced angiotensin AII (AII) regulates the function of many tissues, but the involvement of enterocyte-derived AII in the control of intestinal transport is unknown. This study examined whether there is a local renin-angiotensin system (RAS) in rat villus enterocytes and assessed the effects of AII on SGLT1-dependent glucose transport across the brush border membrane (BBM). Gene and protein expression of angiotensinogen, ACE, and AT(1) and AT(2) receptors were studied in jejunal and ileal enterocytes using immunocytochemistry, Western blotting and RT-PCR. Mucosal uptake of d-[(14)C]glucose by everted intestinal sleeves before and after addition of AII (0-100 nm) to the mucosal buffer was measured in the presence or absence of the AT(1) receptor antagonist losartan (1 microm). Immunocytochemistry revealed the expression of angiotensinogen, ACE, and AT(1) and AT(2) receptors in enterocytes; immunoreactivity of AT(1) receptor and angiotensinogen proteins was especially pronounced at the BBM. Expression of angiotensinogen and AT(1) and AT(2) receptors, but not ACE, was greater in the ileum than the jejunum. Addition of AII to mucosal buffer inhibited phlorizin-sensitive (SGLT1-dependent) jejunal glucose uptake in a rapid and dose-dependent manner and reduced the expression of SGLT1 at the BBM. Losartan attenuated the inhibitory action of AII on glucose uptake. AII did not affect jejunal uptake of l-leucine. The detection of RAS components at the enterocyte BBM, and the rapid inhibition of SGLT1-dependent glucose uptake by luminal AII suggest that AII secretion exerts autocrine control of intestinal glucose transport.


Asunto(s)
Angiotensina II/metabolismo , Enterocitos/metabolismo , Glucosa/metabolismo , Íleon/metabolismo , Yeyuno/metabolismo , Sistema Renina-Angiotensina , Transportador 1 de Sodio-Glucosa/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Animales , Comunicación Autocrina , Western Blotting , Enterocitos/efectos de los fármacos , Íleon/citología , Íleon/efectos de los fármacos , Inmunohistoquímica , Técnicas In Vitro , Yeyuno/citología , Yeyuno/efectos de los fármacos , Leucina/metabolismo , Losartán/farmacología , Masculino , Microvellosidades/metabolismo , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/genética , Receptor de Angiotensina Tipo 2/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/genética
19.
Exp Physiol ; 91(3): 531-7, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16431934

RESUMEN

Previously, it was thought that intestinal phosphate transport occurred exclusively in the proximal small intestine of rodents and humans. However, a recent study has demonstrated that the ileum of mice contributes significantly to the absorption of dietary phosphate, but it is not known whether this region is also an important site of phosphate absorption in the rat. In the present study, we have investigated the mRNA and protein levels of the sodium-phosphate cotransporter, NaPi-IIb, in three regions of rat and mouse small intestine, and related its expression levels to the rate of net phosphate absorption, as measured using the in situ intestinal loop technique. 1,25-Dihydroxyvitamin D3 is an important physiological regulator of intestinal phosphate absorption that increases phosphate transport in both the duodenum and jejunum of the rat. Based on the recently proposed regional profile of phosphate absorption along the mouse small intestine, we have re-evaluated the effects of 1,25-dihydroxyvitamin D3 using three distinct regions of the mouse and rat small intestine. Our studies have revealed important differences in the intestinal handling of phosphate between mice and rats. In mice, maximal phosphate absorption occurs in the ileum, which is paralleled by the highest expression levels of NaPi-IIb mRNA and protein. In contrast, in rats maximal absorption occurs in the duodenum with very little absorption occurring in the ileum, which is similar to the pattern reported in humans. However, in both rodent species only the jejunum shows an increase in phosphate absorption in response to treatment with 1,25-dihydroxyvitamin D3.


Asunto(s)
Absorción Intestinal/efectos de los fármacos , Absorción Intestinal/fisiología , Intestino Delgado/metabolismo , Fosfatos/farmacocinética , Vitamina D/administración & dosificación , Animales , Inyecciones Intraperitoneales , Intestino Delgado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Distribución Tisular
20.
J Physiol ; 553(Pt 1): 137-45, 2003 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12963802

RESUMEN

The mechanism of renal glucose transport involves the reabsorption of filtered glucose from the proximal tubule lumen across the brush border membrane (BBM) via a sodium-dependent transporter, SGLT, and exit across the basolateral membrane via facilitative, GLUT-mediated, transport. The aim of the present study was to determine the effect of streptozotocin-induced diabetes on BBM glucose transport. We found that diabetes increased facilitative glucose transport at the BBM by 67.5 % (P < 0.05)--an effect that was abolished by overnight fasting. Western blotting and immunohistochemistry demonstrated GLUT2 expression at the BBM during diabetes, but the protein was undetectable at the BBM of control animals or diabetic animals that had been fasted overnight. Our findings indicate that streptozotocin-induced diabetes causes the insertion of GLUT2 into the BBM and this may provide a low affinity/high capacity route of entry into proximal tubule cells during hyperglycaemia.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Glucosa/metabolismo , Túbulos Renales Proximales/metabolismo , Proteínas de Transporte de Monosacáridos/biosíntesis , Animales , Glucemia/metabolismo , Western Blotting , Diabetes Mellitus Experimental/patología , Transportador de Glucosa de Tipo 2 , Inmunohistoquímica , Técnicas In Vitro , Corteza Renal/patología , Túbulos Renales Proximales/patología , Cinética , Masculino , Microscopía Confocal , Microvellosidades/metabolismo , Tamaño de los Órganos/fisiología , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA