Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(8)2021 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-33920650

RESUMEN

Electrolytes (NaCl) and fluid malabsorption cause diarrhea in inflammatory bowel disease (IBD). Coupled NaCl absorption, mediated by Na+/H+ and Cl-/HCO3- exchanges on the intestinal villus cells brush border membrane (BBM), is inhibited in IBD. Arachidonic acid metabolites (AAMs) formed via cyclooxygenase (COX) or lipoxygenase (LOX) pathways are elevated in IBD. However, their effects on NaCl absorption are not known. We treated SAMP1/YitFc (SAMP1) mice, a model of spontaneous ileitis resembling human IBD, with Arachidonyl Trifluoro Methylketone (ATMK, AAM inhibitor), or with piroxicam or MK-886, to inhibit COX or LOX pathways, respectively. Cl-/HCO3- exchange, measured as DIDS-sensitive 36Cl uptake, was significantly inhibited in villus cells and BBM vesicles of SAMP1 mice compared to AKR/J controls, an effect reversed by ATMK. Piroxicam, but not MK-886, also reversed the inhibition. Kinetic studies showed that inhibition was secondary to altered Km with no effects on Vmax. Whole cell or BBM protein levels of Down-Regulated in Adenoma (SLC26A3) and putative anion transporter-1 (SLC26A6), the two key BBM Cl-/HCO3- exchangers, were unaltered. Thus, inhibition of villus cell Cl-/HCO3- exchange by COX pathway AAMs, such as prostaglandins, via reducing the affinity of the exchanger for Cl-, and thereby causing NaCl malabsorption, could significantly contribute to IBD-associated diarrhea.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Antiportadores de Cloruro-Bicarbonato/metabolismo , Enterocitos/metabolismo , Ileítis/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Animales , Ácidos Araquidónicos/farmacología , Células Cultivadas , Antiportadores de Cloruro-Bicarbonato/antagonistas & inhibidores , Inhibidores de la Ciclooxigenasa/farmacología , Enterocitos/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Ileítis/genética , Indoles/farmacología , Lipooxigenasa/metabolismo , Inhibidores de la Lipooxigenasa/farmacología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/genética , Piroxicam/farmacología
2.
Int J Mol Sci ; 22(14)2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-34299188

RESUMEN

Na-K-ATPase provides a favorable transcellular Na gradient required for the functioning of Na-dependent nutrient transporters in intestinal epithelial cells. The primary metabolite for enterocytes is glutamine, which is absorbed via Na-glutamine co-transporter (SN2; SLC38A5) in intestinal crypt cells. SN2 activity is stimulated during chronic intestinal inflammation, at least in part, secondarily to the stimulation of Na-K-ATPase activity. Leukotriene D4 (LTD4) is known to be elevated in the mucosa during chronic enteritis, but the way in which it may regulate Na-K-ATPase is not known. In an in vitro model of rat intestinal epithelial cells (IEC-18), Na-K-ATPase activity was significantly stimulated by LTD4. As LTD4 mediates its action via Ca-dependent protein kinase C (PKC), Ca levels were measured and were found to be increased. Phorbol 12-myristate 13-acetate (PMA), an activator of PKC, also mediated stimulation of Na-K-ATPase like LTD4, while BAPTA-AM (Ca chelator) and calphostin-C (Cal-C; PKC inhibitor) prevented the stimulation of Na-K-ATPase activity. LTD4 caused a significant increase in mRNA and plasma membrane protein expression of Na-K-ATPase α1 and ß1 subunits, which was prevented by calphostin-C. These data demonstrate that LTD4 stimulates Na-K-ATPase in intestinal crypt cells secondarily to the transcriptional increase of Na-K-ATPase α1 and ß1 subunits, mediated via the Ca-activated PKC pathway.


Asunto(s)
Calcio/metabolismo , Enteritis/enzimología , Células Epiteliales/enzimología , Intestinos/enzimología , Leucotrieno D4/farmacología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Supervivencia Celular/fisiología , Células Cultivadas , Enteritis/tratamiento farmacológico , Enteritis/patología , Activación Enzimática , Células Epiteliales/efectos de los fármacos , Intestinos/efectos de los fármacos , Proteína Quinasa C/metabolismo , Ratas
3.
Int J Mol Sci ; 22(13)2021 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-34201429

RESUMEN

Obesity increases the risk of postmenopausal breast cancer (BC). This risk is mediated by obesity-induced changes in the adipose-derived secretome (ADS). The pathogenesis of BC in obesity is stimulated by mTOR hyperactivity. In obesity, leucine might support mTOR hyperactivity. Leucine uptake by BC cells is through L-Type Amino Acid Transporter 1 (LAT1). Our objective was to link obesity-ADS induction of LAT1 to the induction of mTOR signaling. Lean- and obese-ADS were obtained from lean and obese mice, respectively. Breast ADS was obtained from BC patients. Estrogen-receptor-positive BC cells were stimulated with ADS. LAT1 activity was determined by uptake of 3H-leucine. The LAT1/CD98 complex, and mTOR signaling were assayed by Western blot. The LAT1 antagonists, BCH and JPH203, were used to inhibit LAT1. Cell migration and invasion were measured by Transwell assays. The results showed obese-ADS-induced LAT1 activity by increasing transporter affinity for leucine. Consistent with this mechanism, LAT1 and CD98 expression were unchanged. Induction of mTOR by obese-ADS was inhibited by LAT1 antagonists. Breast ADS from patients with BMIs > 30 stimulated BC cell migration and invasiveness. Collectively, our findings show that obese-ADS induction of LAT1 supports mTOR hyperactivity in luminal BC cells.


Asunto(s)
Tejido Adiposo/metabolismo , Neoplasias de la Mama/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular , Femenino , Humanos , Leucina/metabolismo , Ratones Endogámicos C57BL , Obesidad/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal
4.
J Nutr ; 150(4): 747-755, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31769840

RESUMEN

BACKGROUND: Chronic alcohol use often leads to malnutrition. However, how the intestinal absorption of nutrients such as glucose may be affected during moderate ethanol use has not been investigated. Glucose is absorbed via sodium (Na)-dependent glucose co-transport (SGLT1; SLC5A1) along the brush border membrane (BBM) of intestinal absorptive villus cells. OBJECTIVE: The aim of this study was to investigate how moderate alcohol consumption affects the absorption of glucose via SGLT1. METHODS: Intestinal epithelial cells (IEC-18; rat) were exposed to 8.64 mM ethanol over 1, 3, 6, and 12 h. Rats (16-wk-old, male, Sprague-Dawley) were administered 2 g/kg ethanol over 1, 3, and 6 h. Na-dependent 3H-O-methyl-d-glucose uptake was measured to assess SGLT1 activity. Na-K-ATPase activity was measured as a function of inorganic phosphate release. Protein expression was analyzed by Western blot analysis and immunohistochemical staining. RESULTS: Ethanol significantly decreased Na-dependent glucose absorption in enterocytes in vitro (ethanol treatment: 48.4% of controls at 1 h; P < 0.01) and in vivo (ethanol treatment: 60.0% of controls at 1 h; P < 0.01). Na-K-ATPase activity was significantly inhibited in vitro (ethanol treatment: 36.9% of controls at 1 h; P < 0.01) and in vivo (ethanol treatment: 42.1% of controls at 1 h; P < 0.01). Kinetic studies showed that the mechanism of inhibition of Na-glucose co-transport was secondary to a decrease in the affinity (1/Km) of the co-transporter for glucose both in vitro and in vivo. Western blots and immunohistochemistry further demonstrated unaltered amounts of SGLT1 after ethanol treatment. CONCLUSIONS: Moderate ethanol significantly decreases glucose absorption in IEC-18 cells and in villus cells of Sprague-Dawley rats. The inhibition of SGLT1 is secondary to an altered Na gradient at the cellular level and secondary to diminished affinity of the co-transporter for glucose at the protein level in the BBM. These observations may, at least in part, explain 1 possible mechanism of the onset of malnutrition associated with alcohol consumption.


Asunto(s)
Células Epiteliales/metabolismo , Etanol/administración & dosificación , Glucosa/metabolismo , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/citología , Transportador 1 de Sodio-Glucosa/antagonistas & inhibidores , Animales , Línea Celular , Células Epiteliales/química , Células Epiteliales/efectos de los fármacos , Intestino Delgado/ultraestructura , Masculino , Microvellosidades/efectos de los fármacos , Microvellosidades/metabolismo , Ratas , Ratas Sprague-Dawley , Sodio/farmacología , Transportador 1 de Sodio-Glucosa/análisis , Transportador 1 de Sodio-Glucosa/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
5.
FASEB J ; 33(8): 9323-9333, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31107610

RESUMEN

During obesity, diabetes and hypertension inevitably coexist and cause innumerable health disparities. In the obesity, diabetes, and hypertension triad (ODHT), deregulation of glucose and NaCl homeostasis, respectively, causes diabetes and hypertension. In the mammalian intestine, glucose is primarily absorbed by Na-glucose cotransport 1 (SGLT1) and coupled NaCl by the dual operation of Na-H exchange 3 (NHE3) and Cl-HCO3 [down-regulated in adenoma (DRA) or putative anion transporter 1 (PAT1)] exchange in the brush border membrane (BBM) of villus cells. The basolateral membrane (BLM) Na/K-ATPase provides the favorable transcellular Na gradient for BBM SGLT1 and NHE3. How these multiple, distinct transport processes may be affected in ODHT is unclear. Here, we show the novel and broad regulation by Na/K-ATPase of glucose and NaCl absorption in ODHT in multiple species (mice, rats, and humans). In vivo, during obesity inhibition of villus-cell BLM, Na/K-ATPase led to compensatory stimulation of BBM SGLT1 and DRA or PAT1, whereas NHE3 was unaffected. Supporting this new cellular adaptive mechanism, direct silencing of BLM Na/K-ATPase in intestinal epithelial cells resulted in selective stimulation of BBM SGLT1 and DRA or PAT1 but not NHE3. These changes will lead to an increase in glucose absorption, maintenance of traditional coupled NaCl absorption, and a de novo increase in NaCl absorption from the novel coupling of stimulated SGLT1 with DRA or PAT1. Thus, these novel observations provide the pathophysiologic basis for the deregulation of glucose and NaCl homeostasis of diabetes and hypertension, respectively, during obesity. These observations may lead to more efficacious treatment for obesity-associated diabetes and hypertension.-Palaniappan, B., Arthur, S., Sundaram, V. L., Butts, M., Sundaram, S., Mani, K., Singh, S., Nepal, N., Sundaram, U. Inhibition of intestinal villus cell Na/K-ATPase mediates altered glucose and NaCl absorption in obesity-associated diabetes and hypertension.


Asunto(s)
Glucosa/metabolismo , Intestinos/citología , Microvellosidades/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Cloruro de Sodio/metabolismo , Animales , Western Blotting , Línea Celular , Técnica del Anticuerpo Fluorescente , Hipertensión/tratamiento farmacológico , Hipertensión/metabolismo , Absorción Intestinal/fisiología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Masculino , Interferencia de ARN , Ratas , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
6.
Int J Mol Sci ; 20(6)2019 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-30917504

RESUMEN

Na-amino acid co-transporters (NaAAcT) are uniquely affected in rabbit intestinal villus cell brush border membrane (BBM) during chronic intestinal inflammation. Specifically, Na-alanine co-transport (ASCT1) is inhibited secondary to a reduction in the affinity of the co-transporter for alanine, whereas Na-glutamine co-transport (B0AT1) is inhibited secondary to a reduction in BBM co-transporter numbers. During chronic intestinal inflammation, there is abundant production of the potent oxidant peroxynitrite (OONO). However, whether OONO mediates the unique alteration in NaAAcT in intestinal epithelial cells during chronic intestinal inflammation is unknown. In this study, ASCT1 and B0AT1 were inhibited by OONO in vitro. The mechanism of inhibition of ASCT1 by OONO was secondary to a reduction in the affinity of the co-transporter for alanine, and secondary to a reduction in the number of co-transporters for B0AT1, which were further confirmed by Western blot analyses. In conclusion, peroxynitrite inhibited both BBM ASCT1 and B0AT1 in intestinal epithelial cells but by different mechanisms. These alterations in the villus cells are similar to those seen in the rabbit model of chronic enteritis. Therefore, this study indicates that peroxynitrite may mediate the inhibition of ASCT1 and B0AT1 during inflammation, when OONO levels are known to be elevated in the mucosa.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Enterocitos/metabolismo , Proteínas de Transporte de Glutamato en la Membrana Plasmática/metabolismo , Microvellosidades/metabolismo , Simportadores/metabolismo , Animales , Línea Celular , Enterocitos/efectos de los fármacos , Enterocitos/patología , Inflamación/metabolismo , Ácido Peroxinitroso/toxicidad , Ratas
7.
J Cell Mol Med ; 22(3): 1443-1451, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29271063

RESUMEN

The only Na-nutrient cotransporter described in mammalian small intestinal crypt cells is SN2/SNAT5, which facilitates glutamine uptake. In a rabbit model of chronic intestinal inflammation, SN2 stimulation is secondary to an increase in affinity of the cotransporter for glutamine. However, the immune regulation of SN2 in the crypt cells during chronic intestinal inflammation is unknown. We sought to determine the mechanism of regulation of Na-nutrient cotransporter SN2 by arachidonic acid metabolites in crypt cells. The small intestines of New Zealand white male rabbits were inflamed via inoculation with Eimeria magna oocytes. After 2-week incubation, control and inflamed rabbits were subjected to intramuscular injections of arachidonyl trifluoromethyl ketone (ATK), piroxicam and MK886 for 48 hrs. After injections, the rabbits were euthanized and crypt cells from small intestines were harvested and used. RESULTS: Treatment of rabbits with ATK prevented the release of AA and reversed stimulation of SN2. Inhibition of cyclooxygenase (COX) with piroxicam did not affect stimulation of SN2. However, inhibition of lipoxygenase (LOX) with MK886, thus reducing leukotriene formation during chronic enteritis, reversed the stimulation of SN2. Kinetic studies showed that the mechanism of restoration of SN2 by ATK or MK886 was secondary to the restoration of the affinity of the cotransporter for glutamine. For all treatment conditions, Western blot analysis revealed no change in SN2 protein levels. COX inhibition proved ineffective at reversing the stimulation of SN2. Thus, this study provides evidence that SN2 stimulation in crypt cells is mediated by the leukotriene pathway during chronic intestinal inflammation.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Ácidos Araquidónicos/farmacología , Coccidiosis/metabolismo , Enteritis/metabolismo , Indoles/farmacología , Inhibidores de la Lipooxigenasa/farmacología , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Ácido Araquidónico/metabolismo , Enfermedad Crónica , Coccidiosis/tratamiento farmacológico , Coccidiosis/parasitología , Coccidiosis/patología , Eimeria/efectos de los fármacos , Eimeria/patogenicidad , Eimeria/fisiología , Enteritis/tratamiento farmacológico , Enteritis/parasitología , Enteritis/patología , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica , Glutamina/metabolismo , Íleon/efectos de los fármacos , Íleon/metabolismo , Íleon/parasitología , Íleon/patología , Leucotrienos/metabolismo , Lipooxigenasa/genética , Lipooxigenasa/metabolismo , Masculino , Prostaglandina-Endoperóxido Sintasas/genética , Prostaglandina-Endoperóxido Sintasas/metabolismo , Conejos , Sodio/metabolismo
8.
Int J Mol Sci ; 19(8)2018 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-30103560

RESUMEN

The progression of cancer is associated with increases in amino acid uptake by cancer cells. Upon their entry into cells through specific transporters, exogenous amino acids are used to synthesize proteins, nucleic acids and lipids and to generate ATP. The essential amino acid leucine is also important for maintaining cancer-associated signaling pathways. By upregulating amino acid transporters, cancer cells gain greater access to exogenous amino acids to support chronic proliferation, maintain metabolic pathways, and to enhance certain signal transduction pathways. Suppressing cancer growth by targeting amino acid transporters will require an in-depth understanding of how cancer cells acquire amino acids, in particular, the transporters involved and which cancer pathways are most sensitive to amino acid deprivation. L-Type Amino Acid Transporter 1 (LAT1) mediates the uptake of essential amino acids and its expression is upregulated during the progression of several cancers. We will review the upstream regulators of LAT1 and the downstream effects caused by the overexpression of LAT1 in cancer cells.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Transportador de Aminoácidos Neutros Grandes 1/biosíntesis , Proteínas de Neoplasias/biosíntesis , Neoplasias/metabolismo , Transducción de Señal , Aminoácidos/metabolismo , Animales , Transporte Biológico Activo , Humanos , Neoplasias/patología
9.
Biochim Biophys Acta ; 1848(2): 702-11, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25462166

RESUMEN

Na-K-ATPase located on the basolateral membrane (BLM) of intestinal epithelial cells provides a favorable intracellular Na+ gradient to promote all Na dependent co-transport processes across the brush border membrane (BBM). Down-regulation of Na-K-ATPase activity has been postulated to alter the absorption via Na-solute co-transporters in human inflammatory bowel disease (IBD). Further, the altered activity of a variety of Na-solute co-transporters in intact villus cells has been reported in animal models of chronic enteritis. But the molecular mechanism of down-regulation of Na-K-ATPase is not known. In the present study, using a rabbit model of chronic intestinal inflammation, which resembles human IBD, Na-K-ATPase in villus cells was shown to decrease. The relative mRNA abundance of α-1 and ß-1 subunits was not altered in villus cells during chronic intestinal inflammation. Similarly, the protein levels of these subunits were also not altered in villus cells during chronic enteritis. However, the BLM concentration of α-1 and ß-1 subunits was diminished in the chronically inflamed intestinal villus cells. An ankyrin-spectrin skeleton is necessary for the proper trafficking of Na-K-ATPase to the BLM of the cell. In the present study, ankyrin expression was markedly diminished in villus cells from the chronically inflamed intestine resulting in depolarization of ankyrin-G protein. The decrease of Na-K-ATPase activity was comparable to that seen in ankyrin knockdown IEC-18 cells. Therefore, altered localization of Na-K-ATPase as a result of transcriptional down-regulation of ankyrin-G mediates the down-regulation of Na-K-ATPase activity during chronic intestinal inflammation.


Asunto(s)
Ancirinas/genética , Membrana Celular/metabolismo , Células Epiteliales/metabolismo , Enfermedades Inflamatorias del Intestino/genética , Microvellosidades/metabolismo , Subunidades de Proteína/genética , ATPasa Intercambiadora de Sodio-Potasio/genética , Actinas/genética , Actinas/metabolismo , Animales , Ancirinas/metabolismo , Membrana Celular/química , Polaridad Celular , Enfermedad Crónica , Modelos Animales de Enfermedad , Células Epiteliales/química , Células Epiteliales/patología , Regulación de la Expresión Génica , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Intestino Delgado/química , Intestino Delgado/metabolismo , Intestino Delgado/patología , Masculino , Microvellosidades/química , Microvellosidades/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Subunidades de Proteína/metabolismo , Conejos , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
10.
Am J Physiol Cell Physiol ; 308(8): C650-6, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25652450

RESUMEN

Na-K-ATPase, an integral membrane protein in mammalian cells, is responsible for maintaining the favorable intracellular Na gradient necessary to promote Na-coupled solute cotransport processes [e.g., Na-glucose cotransport (SGLT1)]. Inhibition of brush border membrane (BBM) SGLT1 is, at least in part, due to the diminished Na-K-ATPase in villus cells from chronically inflamed rabbit intestine. The aim of the present study was to determine the effect of Na-K-ATPase inhibition on the two major BBM Na absorptive pathways, specifically Na-glucose cotransport and Na/H exchange (NHE), in intestinal epithelial (IEC-18) cells. Na-K-ATPase was inhibited using 1 mM ouabain or siRNA for Na-K-ATPase-α1 in IEC-18 cells. SGLT1 activity was determined as 3-O-methyl-D-[(3)H]glucose uptake. Na-K-ATPase activity was measured as the amount of inorganic phosphate released. Treatment with ouabain resulted in SGLT1 inhibition at 1 h but stimulation at 24 h. To further characterize this unexpected stimulation of SGLT1, siRNA silencing was utilized to inhibit Na-K-ATPase-α1. SGLT1 activity was significantly upregulated by Na-K-ATPase silencing, while NHE3 activity remained unaltered. Kinetics showed that the mechanism of stimulation of SGLT1 activity was secondary to an increase in affinity of the cotransporter for glucose without a change in the number of cotransporters. Molecular studies demonstrated that the mechanism of stimulation was not secondary to altered BBM SGLT1 protein levels. Chronic and direct silencing of basolateral Na-K-ATPase uniquely regulates BBM Na absorptive pathways in intestinal epithelial cells. Specifically, while BBM NHE3 is unaffected, SGLT1 is stimulated secondary to enhanced affinity of the cotransporter.


Asunto(s)
Absorción Intestinal/fisiología , Microvellosidades/metabolismo , Transportador 1 de Sodio-Glucosa/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/genética , Transporte Biológico/fisiología , Línea Celular , Coccidiosis/parasitología , Coccidiosis/patología , Eimeria/inmunología , Eimeria/patogenicidad , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Absorción Intestinal/efectos de los fármacos , Absorción Intestinal/genética , Mucosa Intestinal/citología , Mucosa Intestinal/enzimología , Intestino Delgado/citología , Masculino , Ouabaína/farmacología , Interferencia de ARN , ARN Interferente Pequeño , Conejos , Ratas , Sodio/metabolismo , Transportador 1 de Sodio-Glucosa/antagonistas & inhibidores , Transportador 1 de Sodio-Glucosa/genética , Intercambiador 3 de Sodio-Hidrógeno , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , ATPasa Intercambiadora de Sodio-Potasio/genética
11.
Biochim Biophys Acta ; 1838(5): 1208-14, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24412219

RESUMEN

Inhibition of constitutive nitric oxide (cNO) production inhibits SGLT1 activity by a reduction in the affinity for glucose without a change in Vmax in intestinal epithelial cells (IEC-18). Thus, we studied the intracellular pathway responsible for the posttranslational modification/s of SGLT1. NO is known to mediate its effects via cGMP which is diminished tenfold in L-NAME treated cells. Inhibition of cGMP production at the level of guanylyl cyclase or inhibition of protein kinase G also showed reduced SGLT1 activity demonstrating the involvement of PKG pathway in the regulation of SGLT1 activity. Metabolic labeling and immunoprecipitation with anti-SGLT1 specific antibodies did not show any significant changes in phosphorylation of SGLT1 protein. Tunicamycin to inhibit glycosylation reduced SGLT1 activity comparable to that seen with L-NAME treatment. The mechanism of inhibition was secondary to decreased affinity without a change in Vmax. Immunoblots of luminal membranes from tunicamycin treated or L-NAME treated IEC-18 cells showed a decrease in the apparent molecular size of SGLT1 protein to 62 and 67 kD, respectively suggesting an alteration in protein glycosylation. The deglycosylation assay with PNGase-F treatment reduced the apparent molecular size of the specific immunoreactive band of SGLT1 from control and L-NAME treated IEC-18 cells to approximately 62 kD from their original molecular size of 75 kD and 67 kD, respectively. Thus, the posttranslational mechanism responsible for the altered affinity of SGLT1 when cNO is diminished is secondary to altered glycosylation of SGLT1 protein. The intracellular pathway responsible for this alteration is cGMP and its dependent kinase.


Asunto(s)
Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , Procesamiento Proteico-Postraduccional , Transportador 1 de Sodio-Glucosa/genética , Transportador 1 de Sodio-Glucosa/metabolismo , Animales , Células Cultivadas , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Glicosilación , Óxido Nítrico/metabolismo , Fosforilación , Ratas
12.
Nitric Oxide ; 44: 98-104, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25524833

RESUMEN

To facilitate assimilation of glutamine, different Na-dependent glutamine absorptive pathways are present in the rabbit small intestine, specifically B0AT1 in villus and SN2 in crypt cell brush border membrane. Further, both are uniquely regulated in the chronically inflamed intestine. B0AT1 is inhibited secondary to reduced number of brush border membrane (BBM) co-transporters while SN2 is stimulated secondary to an increased affinity for glutamine. These unique changes are reversible by treatment with a broad spectrum immune modulator such as glucocorticoids. However, whether inducible nitric oxide (iNO), known to be elevated in the mucosa of the chronically inflamed intestine, may be responsible for these co-transporter alterations is not known. In the present study, treatment of chronically inflamed rabbits with L-NIL, a selective inhibitor of iNO synthase, reversed the inhibition of B0AT1 in villus and the stimulation of SN2 in crypt cells. At the level of the co-transporter in the brush border membrane, inhibition of iNO production reversed the inhibition of villus B0AT1 by restoring the co-transporter numbers while the stimulation of crypt SN2 was reversed back to normal by restoring its affinity for glutamine. Western blot analyses of BBM proteins also confirmed the kinetic studies. Thus, L-NIL treatment restores the uniquely altered Na-glutamine co-transporters in the enterocytes of chronically inflamed intestine. All these data indicate that iNO functions as an upstream immune modulator directly regulating glutamine assimilation during chronic intestinal inflammation.


Asunto(s)
Gastroenteritis/metabolismo , Glutamina/metabolismo , Íleon/citología , Inflamación/metabolismo , Óxido Nítrico/metabolismo , Animales , Células Cultivadas , Enfermedad Crónica , Cinética , Masculino , Microvellosidades/metabolismo , Microvellosidades/fisiología , Conejos
13.
BMC Gastroenterol ; 15: 47, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25884559

RESUMEN

BACKGROUND: In the chronically inflamed rabbit small intestine, brush border membrane (BBM) Na-glutamine co-transport is inhibited in villus cells (mediated by B0AT1), while it is stimulated in crypt cells (mediated by SN2/SNAT5). How mast cells, known to be enhanced in the chronically inflamed intestine, may regulate B0AT1 in villus and SN2/SNAT5 in crypt cell is unknown. Thus, the aim of the present study is to determine the regulation of B0AT1 and SN2/SNAT5 by mast cells during chronic enteritis. METHODS: Chronic intestinal inflammation was induced in male rabbits with intra-gastric inoculation of Eimeria magna oocytes. Rabbits with chronic inflammation were treated with ketotifen (10 mg/day) or saline (Placebo) for 2 days. Villus and crypts cells were isolated from the rabbit intestine using the Ca++ chelation technique. Na/K-ATPase activity was measured as Pi from cellular homogenate. BBM vesicles (BBMV) were prepared from villus and crypt cells and uptake studies were performed using rapid filtration technique with (3)H-Glutamine. Western blot analyses were done using B0AT1 and SN2 specific antibodies. RESULTS: In villus cells, Na-glutamine co-transport inhibition observed during inflammation was completely reversed by ketotifen, a mast cell stabilizer. In contrast, in crypt cells, Na-glutamine co-transport stimulation was reversed to normal levels by ketotifen. Kinetic studies demonstrated that ketotifen reversed the inhibition of B0AT1 in villus cells by restoring co-transporter numbers in the BBM, whereas the stimulation of SN2/SNAT5 in crypts cells was reversed secondary to restoration of affinity of the co-transporter. Western blot analysis showed that ketotifen restored immune-reactive levels of B0AT1 in villus cells, while SN2/SNAT5 levels from crypts cell remained unchanged. CONCLUSION: In the present study we demonstrate that mast cells likely function as a common upstream immune pathway regulator of the Na-dependent glutamine co-transporters, B0AT1 in villus cells and SN2 in crypts cells that are uniquely altered in the chronically inflamed small intestine.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Degranulación de la Célula/efectos de los fármacos , Enteritis/metabolismo , Glutamina/metabolismo , Mucosa Intestinal/metabolismo , Mastocitos/fisiología , Microvellosidades/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Enfermedad Crónica , Enterocitos/metabolismo , Antagonistas de los Receptores Histamínicos H1/farmacología , Íleon , Mucosa Intestinal/citología , Cetotifen/farmacología , Cinética , Masculino , Mastocitos/enzimología , Microvellosidades/enzimología , Conejos , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , beta-N-Acetilhexosaminidasas/metabolismo
14.
Am J Physiol Cell Physiol ; 307(11): C1010-6, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25231108

RESUMEN

Leukotriene D4 (LTD4) is an important immune inflammatory mediator that is known to be elevated in the mucosa of chronically inflamed intestine and alter nutrient absorption. LTD4 inhibits Na-alanine cotransport in intestinal epithelial cells by decreasing the affinity of the cotransporter ASCT1. LTD4 is known to increase intracellular Ca(++) and cAMP concentrations. However, the intracellular signaling mechanism of LTD4-mediated ASCT1 inhibition is unknown. In the present study, pretreatment with calcium chelator BAPTA-AM or inhibition of Ca(++)-dependent protein kinase C (PKC), specifically PKCα, resulted in the reversal of LTD4-mediated inhibition of ASCT1, revealing the involvement of the Ca(++)-activated PKC pathway. PKCα is known to phosphorylate Raf kinase inhibitor protein (RKIP), thus activating its downstream signaling pathway. Immunoblotting with anti-RKIP-Ser(153) antibody showed an increase in phosphorylation levels of RKIP in LTD4-treated cells. Downregulation of endogenous RKIP showed no decrease in ASCT1 activity by LTD4, thus confirming its involvement in ASCT1 regulation. Phosphorylation of RKIP by PKC is known to activate different signaling pathways, and in this study it was found to activate cAMP-activated protein kinase A (PKA) pathway. Although protein abundance of ASCT1 was not altered in any of the experimental conditions, there was an increase in the levels of phosphothreonine in ASCT1 protein, thus showing that phosphorylation changes were responsible for the altered affinity of ASCT1 by LTD4. In conclusion, LTD4 inhibits ASCT1 through PKC-mediated phosphorylation of RKIP, leading to the subsequent activation of PKA pathway, possibly through ß2-andrenergic receptor activation.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/metabolismo , Células Epiteliales/metabolismo , Mucosa Intestinal/citología , Leucotrieno D4/farmacología , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Proteína Quinasa C/metabolismo , Sistema de Transporte de Aminoácidos ASC/genética , Animales , Western Blotting , Calcio/metabolismo , Línea Celular , Quelantes/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Células Epiteliales/efectos de los fármacos , Microvellosidades , Proteínas de Unión a Fosfatidiletanolamina/genética , Proteína Quinasa C/genética , Ratas
15.
Biochim Biophys Acta ; 1818(3): 434-42, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22100603

RESUMEN

Glutamine is a major nutrient utilized by the intestinal epithelium and is primarily assimilated via Na-glutamine co-transport (NGcT) on the brush border membrane (BBM) of enterocytes. Recently we reported that B(0)AT1 (SLC6A19) mediates glutamine absorption in villus while SN2 (SLC38A5) does the same in crypt cells. However, how B(0)AT1 and SN2 are affected during intestinal inflammation is unknown. In the present study it was shown that during chronic enteritis NGcT was inhibited in villus cells, however, it was stimulated in crypt cells. Our studies also demonstrated that the mechanism of inhibition of NGcT during chronic enteritis was secondary to a reduction in the number of B(0)AT1 co-transporters in the villus cell BBM without a change in the affinity of the co-transporter. In contrast, stimulation of NGcT in crypt cells was secondary to an increase in the affinity of SN2 for glutamine without an alteration in the number of co-transporters. Thus, glutamine assimilation which occurs via distinct transporters in crypt and villus cells is altered in the chronically inflamed intestine.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Enteritis/metabolismo , Glutamina/metabolismo , Mucosa Intestinal/metabolismo , Animales , Transporte Biológico , Enfermedad Crónica , Enteritis/patología , Inflamación/metabolismo , Inflamación/patología , Mucosa Intestinal/patología , Masculino , Conejos
16.
PLoS One ; 18(11): e0294387, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37943850

RESUMEN

[This corrects the article DOI: 10.1371/journal.pone.0203552.].

17.
Life (Basel) ; 13(9)2023 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-37763336

RESUMEN

INTRODUCTION: Metabolic syndrome amplifies the risk of gestational diabetes, preeclampsia, and preterm labor in pregnant women. Similarly, women with substance use disorder have worsened obstetric and birth outcomes. Despite these two conditions being major healthcare disparities in Appalachia, the health outcomes of this cohort have not been studied thus far. This study looks at the health outcomes of this cohort. METHOD AND RESULTS: In this retrospective cohort study, we analyzed 27,955 mothers who delivered at Cabell Huntington Hospital between January 2010 and November 2021. We implemented Chi-square tests to determine the associations and multiple logistic regression methods for comparison after controlling for other factors, and found that MetS, together with SUD, significantly increases the risk as well as the number of pregnancy complications such as gestational diabetes (p-value < 0.001), preeclampsia (p-value < 0.001), premature rupture (p-value < 0.001), preterm labor (p-value < 0.001), and newborn disorder (p-value < 0.001) compared to the women who had none or had either MetS or SUD alone. CONCLUSION: Women with both metabolic syndrome and substance abuse had worsened pregnancy and neonatal outcomes compared to women with metabolic syndrome or SUD alone. In conclusion, analysis of all the variables is crucial to strategically planning and implementing health interventions that will positively influence the health outcome of the pregnant woman as well as the child.

18.
Nutrients ; 14(3)2022 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-35276983

RESUMEN

The gut microbiota is a complex community of microorganisms that has become a new focus of attention due to its association with numerous human diseases. Research over the last few decades has shown that the gut microbiota plays a considerable role in regulating intestinal homeostasis, and disruption to the microbial community has been linked to chronic disease conditions such as inflammatory bowel disease (IBD), colorectal cancer (CRC), and obesity. Obesity has become a global pandemic, and its prevalence is increasing worldwide mostly in Western countries due to a sedentary lifestyle and consumption of high-fat/high-sugar diets. Obesity-mediated gut microbiota alterations have been associated with the development of IBD and IBD-induced CRC. This review highlights how obesity-associated dysbiosis can lead to the pathogenesis of IBD and CRC with a special focus on mechanisms of altered absorption of short-chain fatty acids (SCFAs).


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Enfermedad Crónica , Disbiosis/complicaciones , Microbioma Gastrointestinal/fisiología , Humanos , Enfermedades Inflamatorias del Intestino/complicaciones , Obesidad/complicaciones
19.
Inflamm Bowel Dis ; 27(11): 1804-1812, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34019094

RESUMEN

In the small intestine, Na:H (NHE3) and Cl:HCO3 (DRA or PAT1) exchangers present in the brush border membrane (BBM) of absorptive villus cells are primarily responsible for the coupled absorption of NaCl, the malabsorption of which causes diarrhea, a common symptom of inflammatory bowel disease (IBD). Inducible nitric oxide (iNO), a known mediator of inflammation, is increased in the mucosa of the chronically inflamed IBD intestine. An SAMP1/YitFc (SAMP1) mouse, a spontaneous model of chronic ileitis very similar to human IBD, was used to study alterations in NaCl absorption. The SAMP1 and control AKR mice were treated with I-N(6)-(1-Iminoethyl)-lysine (L-NIL) to inhibit iNO production, and DRA/PAT1 and NHE3 activities and protein expression were studied. Though Na:H exchange activity was unaffected, Cl:HCO3 activity was significantly decreased in SAMP1 mice due to a reduction in its affinity for Cl, which was reversed by L-NIL treatment. Though DRA and PAT1 expressions were unchanged in all experimental conditions, phosphorylation studies indicated that DRA, not PAT1, is affected in SAMP1. Moreover, the altered phosphorylation levels of DRA was restored by L-NIL treatment. Inducible NO mediates the inhibition of coupled NaCl absorption by decreasing Cl:HCO3 but not Na:H exchange. Specifically, Cl:HCO3 exchanger DRA but not PAT1 is regulated at the level of its phosphorylation by iNO in the chronically inflamed intestine.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Óxido Nítrico/metabolismo , Cloruro de Sodio , Intercambiadores de Sodio-Hidrógeno , Sistemas de Transporte de Aminoácidos , Animales , Antiportadores , Modelos Animales de Enfermedad , Inflamación , Enfermedades Inflamatorias del Intestino/metabolismo , Absorción Intestinal , Intestinos , Ratones , Cloruro de Sodio/metabolismo , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/metabolismo , Transportadores de Sulfato , Simportadores
20.
Cells ; 10(4)2021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33805551

RESUMEN

The primary means of intestinal absorption of nutrients by villus cells is via Na-dependent nutrient co-transporters located in the brush border membrane (BBM). These secondary active co-transport processes require a favorable transcellular Na gradient that is provided by Na-K-ATPase. In chronic enteritis, malabsorption of essential nutrients is partially due to inhibition of villus Na-K-ATPase activity mediated by specific immune inflammatory mediators that are known to be elevated in the inflamed mucosa. However, how Prostaglandin E2 (PGE2), a specific mediator of nutrient malabsorption in the villus BBM, may mediate the inhibition of Na-K-ATPase is not known. Therefore, this study aimed to determine the effect of PGE2 on Na-K-ATPase in villus cells and define its mechanism of action. In vitro, in IEC-18 cells, PGE2 treatment significantly reduced Na-K-ATPase activity, accompanied by a significant increase in the intracellular levels of cyclic Adenosine Monophosphate (cAMP). The treatment with cAMP analog 8-Bromo-cAMP mimicked the PGE2-mediated effect on Na-K-ATPase activity, while Rp-cAMP (PKA inhibitor) pretreatment reversed the same. The mechanism of inhibition of PGE2 was secondary to a transcriptional reduction in the Na-K-ATPase α1 and ß1 subunit genes, which was reversed by the Rp-cAMP pretreatment. Thus, the PGE2-mediated activation of the PKA pathway mediates the transcriptional inhibition of Na-K-ATPase activity in vitro.


Asunto(s)
Dinoprostona/farmacología , Células Epiteliales/enzimología , Intestinos/citología , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Epiteliales/efectos de los fármacos , Espacio Intracelular/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores de Prostaglandina/antagonistas & inhibidores , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/genética , Xantonas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA