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










Base de datos
Intervalo de año de publicación
1.
Am J Physiol Endocrinol Metab ; 325(5): E581-E594, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37819196

RESUMEN

Male mice lacking the Na+-K+-2Cl- cotransporter Slc12a2 (Nkcc1) specifically in insulin-secreting ß-cells (Slc12a2ßKO) have reduced ß-cell mass and mild ß-cell secretory dysfunction associated with overweight, glucose intolerance, insulin resistance, and metabolic abnormalities. Here, we confirmed and extended previous results to female Slc12a2ßKO mice, which developed a similar metabolic syndrome-like phenotype as males, albeit milder. Notably, male and female Slc12a2ßKO mice developed overweight without consuming excess calories. Analysis of the feeding microstructure revealed that young lean Slc12a2ßKO male mice ate meals of higher caloric content and at a relatively lower frequency than normal mice, particularly during the night. In addition, overweight Slc12a2ßKO mice consumed significantly larger meals than lean mice. Therefore, the reduced satiation control of feeding precedes the onset of overweight and is worsened in older Slc12a2ßKO mice. However, the time spent between meals remained intact in lean and overweight Slc12a2ßKO mice, indicating conserved satiety responses to ad libitum feeding. Nevertheless, satiety was intensified during and after refeeding only in overweight males. In lean females, satiety responses to refeeding were delayed relative to age- and body weight-matched control mice but normalized in overweight mice. Since meal size did not change during refeeding, these data suggested that the satiety control of eating after fasting is impaired in lean Slc12a2ßKO mice before the onset of overweight and independently of their reduced satiation responses. Therefore, our results support the novel hypothesis that reduced satiation precedes the onset of overweight and the development of metabolic dysregulation.NEW & NOTEWORTHY Obesity, defined as excess fat accumulation, increases the absolute risk for metabolic diseases. Although obesity is usually attributed to increased food intake, we demonstrate that body weight gain can be hastened without consuming excess calories. In fact, impaired meal termination control, i.e., satiation, is detectable before the development of overweight in an animal model that develops a metabolic syndrome-like phenotype.


Asunto(s)
Insulinas , Síndrome Metabólico , Masculino , Femenino , Ratones , Animales , Sobrepeso/genética , Saciedad , Obesidad/genética , Ingestión de Energía , Insulina/metabolismo
2.
PLoS One ; 17(12): e0279560, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36580474

RESUMEN

The risk of type-2 diabetes and cardiovascular disease is higher in subjects with metabolic syndrome, a cluster of clinical conditions characterized by obesity, impaired glucose metabolism, hyperinsulinemia, hyperlipidemia and hypertension. Diuretics are frequently used to treat hypertension in these patients, however, their use has long been associated with poor metabolic outcomes which cannot be fully explained by their diuretic effects. Here, we show that mice lacking the diuretic-sensitive Na+K+2Cl-cotransporter-1 Nkcc1 (Slc12a2) in insulin-secreting ß-cells of the pancreatic islet (Nkcc1ßKO) have reduced in vitro insulin responses to glucose. This is associated with islet hypoplasia at the expense of fewer and smaller ß-cells. Remarkably, Nkcc1ßKO mice excessively gain weight and progressive metabolic syndrome when fed a standard chow diet ad libitum. This is characterized by impaired hepatic insulin receptor activation and altered lipid metabolism. Indeed, overweight Nkcc1ßKO but not lean mice had fasting and fed hyperglycemia, hypertriglyceridemia and non-alcoholic steatohepatitis. Notably, fasting hyperinsulinemia was detected earlier than hyperglycemia, insulin resistance, glucose intolerance and increased hepatic de novo gluconeogenesis. Therefore, our data provide evidence supporting the novel hypothesis that primary ß-cell defects related to Nkcc1-regulated intracellular Cl-homeostasis and ß-cell growth can result in the development of metabolic syndrome shedding light into additional potential mechanisms whereby chronic diuretic use may have adverse effects on metabolic homeostasis in susceptible individuals.


Asunto(s)
Hiperglucemia , Hiperinsulinismo , Hipertensión , Resistencia a la Insulina , Células Secretoras de Insulina , Síndrome Metabólico , Ratones , Animales , Síndrome Metabólico/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Hiperinsulinismo/metabolismo , Hiperglucemia/metabolismo , Diuréticos , Hipertensión/metabolismo
3.
PLoS One ; 16(2): e0246569, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33539467

RESUMEN

The feeding pattern and control of energy intake in mice housed in groups are poorly understood. Here, we determined and quantified the normal feeding microstructure of social male and female mice of the C57BL/6J genetic background fed a chow diet. Mice at 10w, 20w and 30w of age showed the expected increase in lean and fat mass, being the latter more pronounced and variable in males than in females. Under ad libitum conditions, 20w and 30w old females housed in groups showed significantly increased daily energy intake when adjusted to body weight relative to age-matched males. This was the combined result of small increases in energy intake during the nocturnal and diurnal photoperiods of the day without major changes in the circadian pattern of energy intake or spontaneous ambulatory activity. The analysis of the feeding microstructure suggests sex- and age-related contributions of meal size, meal frequency and intermeal interval to the control of energy intake under stable energy balance, but not under negative energy balance imposed by prolonged fasting. During the night, 10-20w old females ate less frequently bigger meals and spent more time eating them resulting in reduced net energy intake relative to age-matched males. In addition, male and female mice at all ages tested significantly shortened the intermeal interval during the first hours of re-feeding in response to fasting without affecting meal size. Further, 20-30w old males lengthened their intermeal interval as re-feeding time increased to reach fed-levels faster than age-matched females. Collectively, our results suggest that the physiological mechanisms controlling meal size (satiation) and the non-eating time spent between meals (satiety) during stable or negative energy balance are regulated in a sex- and age-dependent manner in social mice.


Asunto(s)
Ingestión de Energía/fisiología , Conducta Alimentaria/fisiología , Animales , Peso Corporal/fisiología , Grasas de la Dieta , Metabolismo Energético/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Saciedad/fisiología
4.
PLoS One ; 15(12): e0242749, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33264332

RESUMEN

Cystic fibrosis (CF) is due to mutations in the CF-transmembrane conductance regulator (CFTR) and CF-related diabetes (CFRD) is its most common co-morbidity, affecting ~50% of all CF patients, significantly influencing pulmonary function and longevity. Yet, the complex pathogenesis of CFRD remains unclear. Two non-mutually exclusive underlying mechanisms have been proposed in CFRD: i) damage of the endocrine cells secondary to the severe exocrine pancreatic pathology and ii) intrinsic ß-cell impairment of the secretory response in combination with other factors. The later has proven difficult to determine due to low expression of CFTR in ß-cells, which results in the general perception that this Cl-channel does not participate in the modulation of insulin secretion or the development of CFRD. The objective of the present work is to demonstrate CFTR expression at the molecular and functional levels in insulin-secreting ß-cells in normal human islets, where it seems to play a role. Towards this end, we have used immunofluorescence confocal and immunofluorescence microscopy, immunohistochemistry, RT-qPCR, Western blotting, pharmacology, electrophysiology and insulin secretory studies in normal human, rat and mouse islets. Our results demonstrate heterogeneous CFTR expression in human, mouse and rat ß-cells and provide evidence that pharmacological inhibition of CFTR influences basal and stimulated insulin secretion in normal mouse islets but not in islets lacking this channel, despite being detected by electrophysiological means in ~30% of ß-cells. Therefore, our results demonstrate a potential role for CFTR in the pancreatic ß-cell secretory response suggesting that intrinsic ß-cell dysfunction may also participate in the pathogenesis of CFRD.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Secretoras de Insulina/metabolismo , Adulto , Anciano , Animales , Anticuerpos/metabolismo , Antígenos/metabolismo , Línea Celular , Regulador de Conductancia de Transmembrana de Fibrosis Quística/inmunología , Femenino , Humanos , Lactante , Secreción de Insulina , Masculino , Ratones , Persona de Mediana Edad , Ratas , Reproducibilidad de los Resultados , Adulto Joven
5.
Biochem Soc Trans ; 47(6): 1843-1855, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31697318

RESUMEN

It is accepted that insulin-secreting ß-cells release insulin in response to glucose even in the absence of functional ATP-sensitive K+ (KATP)-channels, which play a central role in a 'consensus model' of secretion broadly accepted and widely reproduced in textbooks. A major shortcoming of this consensus model is that it ignores any and all anionic mechanisms, known for more than 40 years, to modulate ß-cell electrical activity and therefore insulin secretion. It is now clear that, in addition to metabolically regulated KATP-channels, ß-cells are equipped with volume-regulated anion (Cl-) channels (VRAC) responsive to glucose concentrations in the range known to promote electrical activity and insulin secretion. In this context, the electrogenic efflux of Cl- through VRAC and other Cl- channels known to be expressed in ß-cells results in depolarization because of an outwardly directed Cl- gradient established, maintained and regulated by the balance between Cl- transporters and channels. This review will provide a succinct historical perspective on the development of a complex hypothesis: Cl- transporters and channels modulate insulin secretion in response to nutrients.


Asunto(s)
Cloruros/metabolismo , Células Secretoras de Insulina/fisiología , Canales Iónicos/metabolismo , Modelos Biológicos , Animales , Humanos , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Transporte Iónico
6.
Am J Physiol Cell Physiol ; 317(4): C843-C856, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31365295

RESUMEN

The Na+K+2Cl- cotransporter-2 (Nkcc2, Slc12a1) is abundantly expressed in the kidney and its inhibition with the loop-diuretics bumetanide and furosemide has been linked to transient or permanent hyperglycemia in mice and humans. Notably, Slc12a1 is expressed at low levels in hypothalamic neurons and in insulin-secreting ß-cells of the endocrine pancreas. The present study was designed to determine if global elimination of one of the Slc12a1 products, i.e., Nkcc2 variant a (Nkcc2a), the main splice version of Nkcc2 found in insulin-secreting ß-cells, has an impact on the insulin and glucagon secretory responses and fuel homeostasis in vivo. We have used dynamic tests of glucose homeostasis in wild-type mice and mice lacking both alleles of Nkcc2a (Nkcc2aKO) and assessed their islet secretory responses in vitro. Under basal conditions, Nkcc2aKO mice have impaired glucose homeostasis characterized by increased blood glucose, intolerance to the sugar, delayed/blunted in vivo insulin and glucagon responses to glucose, and increased glycemic responses to the gluconeogenic substrate alanine. Further, we provide evidence of conserved quantitative secretory responses of Nkcc2aKO islets within a context of increased islet size related to hyperplastic/hypertrophic glucagon- and insulin-positive cells (α-cells and ß-cells, respectively), normal total islet Cl- content, and reduced ß-cell expression of the Cl- extruder Kcc2.


Asunto(s)
Glucagón/metabolismo , Glucosa/metabolismo , Insulina/farmacología , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Animales , Intolerancia a la Glucosa/tratamiento farmacológico , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones Noqueados , Ratones Transgénicos , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología
7.
Sci Rep ; 7(1): 1732, 2017 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-28496181

RESUMEN

Intracellular chloride concentration ([Cl-]i) in pancreatic ß-cells is kept above electrochemical equilibrium due to the predominant functional presence of Cl- loaders such as the Na+K+2Cl- co-transporter 1 (Slc12a2) over Cl-extruders of unidentified nature. Using molecular cloning, RT-PCR, Western blotting, immunolocalization and in vitro functional assays, we establish that the "neuron-specific" K+Cl- co-transporter 2 (KCC2, Slc12a5) is expressed in several endocrine cells of the pancreatic islet, including glucagon secreting α-cells, but particularly in insulin-secreting ß-cells, where we provide evidence for its role in the insulin secretory response. Three KCC2 splice variants were identified: the formerly described KCC2a and KCC2b along with a novel one lacking exon 25 (KCC2a-S25). This new variant is undetectable in brain or spinal cord, the only and most abundant known sources of KCC2. Inhibition of KCC2 activity in clonal MIN6 ß-cells increases basal and glucose-stimulated insulin secretion and Ca2+ uptake in the presence of glibenclamide, an inhibitor of the ATP-dependent potassium (KATP)-channels, thus suggesting a possible mechanism underlying KCC2-dependent insulin release. We propose that the long-time considered "neuron-specific" KCC2 co-transporter is expressed in pancreatic islet ß-cells where it modulates Ca2+-dependent insulin secretion.


Asunto(s)
Secreción de Insulina , Neuronas/metabolismo , Simportadores/metabolismo , Empalme Alternativo/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Calcio/metabolismo , Línea Celular , Glucosa/farmacología , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Canales KATP/metabolismo , Ratones , Piridazinas , Simportadores/química , Simportadores/genética , Tiazoles , Cotransportadores de K Cl
8.
Lab Anim ; 51(2): 138-146, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27166392

RESUMEN

Genotyping of genetically-engineered mice is necessary for the effective design of breeding strategies and identification of mutant mice. This process relies on the identification of DNA markers introduced into genomic sequences of mice, a task usually performed using the polymerase chain reaction (PCR). Clearly, the limiting step in genotyping is isolating pure genomic DNA. Isolation of mouse DNA for genotyping typically involves painful procedures such as tail snip, digit removal, or ear punch. Although the harvesting of hair has previously been proposed as a source of genomic DNA, there has been a perceived complication and reluctance to use this non-painful technique because of low DNA yields and fear of contamination. In this study we developed a simple, economic, and efficient strategy using Chelex® resins to purify genomic DNA from hair roots of mice which are suitable for genotyping. Upon comparison with standard DNA purification methods using a commercially available kit, we demonstrate that Chelex® efficiently and consistently purifies high-quality DNA from hair roots, minimizing pain, shortening time and reducing costs associated with the determination of accurate genotypes. Therefore, the use of hair roots combined with Chelex® is a reliable and more humane alternative for DNA genotyping.


Asunto(s)
Quelantes/química , ADN/aislamiento & purificación , Técnicas de Genotipaje/métodos , Folículo Piloso/química , Ratones/genética , Poliestirenos/química , Polivinilos/química , Animales , Animales Modificados Genéticamente/genética , Femenino , Masculino
9.
Front Cell Dev Biol ; 4: 150, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28101499

RESUMEN

Na+K+2Cl- co-transporters (NKCCs) effect the electroneutral movement of Na+-K+ and 2Cl- ions across the plasma membrane of vertebrate cells. There are two known NKCC isoforms, NKCC1 (Slc12a2) and NKCC2 (Slc12a1). NKCC1 is a ubiquitously expressed transporter involved in cell volume regulation, Cl- homeostasis and epithelial salt secretion, whereas NKCC2 is abundantly expressed in kidney epithelial cells of the thick ascending loop of Henle, where it plays key roles in NaCl reabsorption and electrolyte homeostasis. Although NKCC1 and NKCC2 co-transport the same ions with identical stoichiometry, NKCC1 actively co-transports water whereas NKCC2 does not. There is growing evidence showing that NKCC2 is expressed outside the kidney, but its function in extra-renal tissues remains unknown. The present study shows molecular and functional evidence of endogenous NKCC2 expression in COS7 cells, a widely used mammalian cell model. Endogenous NKCC2 is primarily found in recycling endosomes, Golgi cisternae, Golgi-derived vesicles, and to a lesser extent in the endoplasmic reticulum. Unlike NKCC1, NKCC2 is minimally hybrid/complex N-glycosylated under basal conditions and yet it is trafficked to the plasma membrane region of hyper-osmotically challenged cells through mechanisms that require minimal complex N-glycosylation or functional Golgi cisternae. Control COS7 cells exposed to slightly hyperosmotic (~6.7%) solutions for 16 h were not shrunken, suggesting that either one or both NKCC1 and NKCC2 may participate in cell volume recovery. However, NKCC2 targeted to the plasma membrane region or transient over-expression of NKCC2 failed to rescue NKCC1 in COS7 cells where NKCC1 had been silenced. Further, COS7 cells in which NKCC1, but not NKCC2, was silenced exhibited reduced cell size compared to control cells. Altogether, these results suggest that NKCC2 does not participate in cell volume recovery and therefore, NKCC1 and NKCC2 are functionally different Na+K+2Cl- co-transporters.

10.
Int J Cell Biol ; 2015: 505294, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26351455

RESUMEN

The Na(+)K(+)2Cl(-) cotransporter-1 (Slc12a2, NKCC1) is widely distributed and involved in cell volume/ion regulation. Functional NKCC1 locates in the plasma membrane of all cells studied, particularly in the basolateral membrane of most polarized cells. Although the mechanisms involved in plasma membrane sorting of NKCC1 are poorly understood, it is assumed that N-glycosylation is necessary. Here, we characterize expression, N-glycosylation, and distribution of NKCC1 in COS7 cells. We show that ~25% of NKCC1 is complex N-glycosylated whereas the rest of it corresponds to core/high-mannose and hybrid-type N-glycosylated forms. Further, ~10% of NKCC1 reaches the plasma membrane, mostly as core/high-mannose type, whereas ~90% of NKCC1 is distributed in defined intracellular compartments. In addition, inhibition of the first step of N-glycan biosynthesis with tunicamycin decreases total and plasma membrane located NKCC1 resulting in almost undetectable cotransport function. Moreover, inhibition of N-glycan maturation with swainsonine or kifunensine increased core/hybrid-type NKCC1 expression but eliminated plasma membrane complex N-glycosylated NKCC1 and transport function. Together, these results suggest that (i) NKCC1 is delivered to the plasma membrane of COS7 cells independently of its N-glycan nature, (ii) most of NKCC1 in the plasma membrane is core/hybrid-type N-glycosylated, and (iii) the minimal proportion of complex N-glycosylated NKCC1 is functionally active.

11.
J Endocrinol ; 227(3): 153-65, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26400961

RESUMEN

The products of the Slc12a1 and Slc12a2 genes, commonly known as Na(+)-dependent K(+)2Cl(-) co-transporters NKCC2 and NKCC1, respectively, are the targets for the diuretic bumetanide. NKCCs are implicated in the regulation of intracellular chloride concentration ([Cl(-)]i) in pancreatic ß-cells, and as such, they may play a role in glucose-stimulated plasma membrane depolarization and insulin secretion. Unexpectedly, permanent elimination of NKCC1 does not preclude insulin secretion, an event potentially linked to the homeostatic regulation of additional Cl(-) transporters expressed in ß-cells. In this report we provide evidence for such a mechanism. Mice lacking a single allele of Slc12a2 exhibit lower fasting glycemia, increased acute insulin response (AIR) and lower blood glucose levels 15-30 min after a glucose load when compared to mice harboring both alleles of the gene. Furthermore, heterozygous expression or complete absence of Slc12a2 associates with increased NKCC2 protein expression in rodent pancreatic ß-cells. This has been confirmed by using chronic pharmacological down-regulation of NKCC1 with bumetanide in the mouse MIN6 ß-cell line or permanent molecular silencing of NKCC1 in COS7 cells, which results in increased NKCC2 expression. Furthermore, MIN6 cells chronically pretreated with bumetanide exhibit increased initial rates of Cl(-) uptake while preserving glucose-stimulated insulin secretion. Together, our results suggest that NKCCs are involved in insulin secretion and that a single Slc12a2 allele may protect ß-cells from failure due to increased homeostatic expression of Slc12a1.


Asunto(s)
Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Animales , Bumetanida/farmacología , Regulación hacia Abajo/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Heterocigoto , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Ratones Transgénicos , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología , Miembro 1 de la Familia de Transportadores de Soluto 12/genética , Miembro 2 de la Familia de Transportadores de Soluto 12/genética
12.
Am J Physiol Cell Physiol ; 308(9): C767-77, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25740155

RESUMEN

The renin angiotensin system (RAS) plays a vital role in the regulation of the cardiovascular and renal functions. COS7 is a robust and easily transfectable cell line derived from the kidney of the African green monkey, Cercopithecus aethiops. The aims of this study were to 1) demonstrate the presence of an endogenous and functional RAS in COS7, and 2) investigate the role of a disintegrin and metalloproteinase-17 (ADAM17) in the ectodomain shedding of angiotensin converting enzyme-2 (ACE2). Reverse transcription coupled to gene-specific polymerase chain reaction demonstrated expression of ACE, ACE2, angiotensin II type 1 receptor (AT1R), and renin at the transcript levels in total RNA cell extracts. Western blot and immunohistochemistry identified ACE (60 kDa), ACE2 (75 kDa), AT1R (43 kDa), renin (41 kDa), and ADAM17 (130 kDa) in COS7. At the functional level, a sensitive and selective mass spectrometric approach detected endogenous renin, ACE, and ACE2 activities. ANG-(1-7) formation (m/z 899) from the natural substrate ANG II (m/z 1,046) was detected in lysates and media. COS7 cells stably expressing shRNA constructs directed against endogenous ADAM17 showed reduced ACE2 shedding into the media. This is the first study demonstrating endogenous expression of the RAS and ADAM17 in the widely used COS7 cell line and its utility to study ectodomain shedding of ACE2 mediated by ADAM17 in vitro. The transfectable nature of this cell line makes it an attractive cell model for studying the molecular, functional, and pharmacological properties of the renal RAS.


Asunto(s)
Proteínas ADAM/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Sistema Renina-Angiotensina , Proteínas ADAM/genética , Proteína ADAM17 , Angiotensina I/metabolismo , Angiotensina II/genética , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Secuencia de Bases , Células COS , Chlorocebus aethiops , Datos de Secuencia Molecular , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/genética , Interferencia de ARN , ARN Mensajero/metabolismo , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Renina/genética , Renina/metabolismo , Sistema Renina-Angiotensina/genética , Transcripción Genética , Transfección
13.
Cell Physiol Biochem ; 32(7): 14-31, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24429812

RESUMEN

The homologous genes encoding the electroneutral solute carrier family 12A (SLC12A) were identified more than 20 years ago, however, over the last few years, it has become clear that each of the genes within this family potentially encode for more than one cation-chloride cotransporter (CCC). Even more surprising, despite more than 30 years of functional studies and a wealth of knowledge on the activators, inhibitors, ion affinities, and kinetics of these cotransporters, we still cannot sufficiently explain why some cells express only one CCC isoform, while others express two, three, or more CCC isoforms. In 2009, Drs. Alvarez-Leefmans and Di Fulvio published an extensive in silico molecular analysis of the potential splice variants of the Na(+)-dependent cation-chloride cotransporters. In this review, we will look at the exceptionally large variety of potential splice variants within the Na(+)-independent cation-chloride cotransporter (SLC12A4-SLC12A7) genes, their initial tissue identification, and their physiological relevance.


Asunto(s)
Cloruros/metabolismo , Transporte Iónico/genética , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Miembro 4 de la Familia de Transportadores de Soluto 12/metabolismo , Simportadores/metabolismo , Proteínas Portadoras/química , Cationes/metabolismo , Humanos , Cinética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Simportadores de Cloruro de Sodio-Potasio/química , Simportadores de Cloruro de Sodio-Potasio/genética , Miembro 4 de la Familia de Transportadores de Soluto 12/química , Miembro 4 de la Familia de Transportadores de Soluto 12/genética , Simportadores/química , Simportadores/genética
14.
J Endocrinol ; 215(1): 59-70, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22872759

RESUMEN

The intracellular chloride concentration ([Cl(-)](i)) in ß-cells plays an important role in glucose-stimulated plasma membrane depolarisation and insulin secretion. [Cl(-)](i) is maintained above equilibrium in ß-cells by the action of Cl(-) co-transporters of the solute carrier family 12 group A (Slc12a). ß-Cells express Slc12a1 and Slc12a2, which are known as the bumetanide (BTD)-sensitive Na(+)-dependent K(+)2Cl(-) co-transporters 2 and 1 respectively. We show that mice lacking functional alleles of the Slc12a2 gene exhibit better fasting glycaemia, increased insulin secretion in response to glucose, and improved glucose tolerance when compared with wild-type (WT). This phenomenon correlated with increased sensitivity of ß-cells to glucose in vitro and with increased ß-cell mass. Further, administration of low doses of BTD to mice deficient in Slc12a2 worsened their glucose tolerance, and low concentrations of BTD directly inhibited glucose-stimulated insulin secretion from ß-cells deficient in Slc12a2 but expressing intact Slc12a1 genes. Together, our results suggest for the first time that the Slc12a2 gene is not necessary for insulin secretion and that its absence increases ß-cell secretory capacity. Further, impairment of insulin secretion with BTD in vivo and in vitro in islets lacking Slc12a2 genes unmasks a potential new role for Slc12a1 in ß-cell physiology.


Asunto(s)
Silenciador del Gen , Intolerancia a la Glucosa/genética , Insulina/metabolismo , Simportadores de Cloruro de Sodio-Potasio/genética , Animales , Células Cultivadas , Femenino , Silenciador del Gen/fisiología , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Homocigoto , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Simportadores de Cloruro de Sodio-Potasio/fisiología , Miembro 2 de la Familia de Transportadores de Soluto 12 , Regulación hacia Arriba
15.
Cell Physiol Biochem ; 30(1): 95-112, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22759959

RESUMEN

The solute carrier protein family 12 group A, member one (Slc12a1) and two (Slc12a2) encode several splice variants of the kidney-specific and the ubiquitous isoforms, respectively, of the bumetanide (BTD)-sensitive Na-dependent K2Cl co-transporter. The Slc12a2 co-transporter is involved in the maintenance of a high intracellular chloride concentration [Cl(-)](i) in ß-cells and its inhibition with BTD blocks glucose-induced insulin secretion. In ß-cells, [Cl(-)](i) plays an important role in glucose-induced depolarization and insulin secretion. Glucose promotes electrogenic efflux of Cl(-) contributing to ß-cell's electrical and secretory activity. To identify the expression pattern of Slc12a1 and Slc12a2 genes in ß-cells we have used RT-PCR, Western blotting and immunolocalization studies in mouse pancreatic islets, ß-cell lines and rat tissues. Our results demonstrate expression of specific splice variants of Slc12a1 and Slc12a2 transcripts in ß-cells i.e., variants 1 of Slc12a1 (NKCC2A) and Slc12a2 (NKCC1a). Molecular cloning and characterization of Slc12a1 variant 1 transcripts from ß-cells revealed an alternative splicing event involving the 5'-UTR region. NKCC2A expression at the protein level in islets and ß-cells was confirmed by immunoblotting and immunolocalization. Further, NKCC2A, NKCC1a and pro-insulin co-localized in ß-cells but not in the exocrine pancreas. Therefore, our results provide for the first time evidence of NKCC2A expression in pancreatic ß-cells where it may play a role in insulin secretion.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Etiquetas de Secuencia Expresada , Expresión Génica , Insulina/genética , Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Especificidad de Órganos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Alineación de Secuencia , Análisis de Secuencia de ADN , Simportadores de Cloruro de Sodio-Potasio/genética , Miembro 1 de la Familia de Transportadores de Soluto 12 , Miembro 2 de la Familia de Transportadores de Soluto 12
16.
Cell Physiol Biochem ; 29(1-2): 303-12, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22415099

RESUMEN

The fetal human lens epithelial cell (LEC) line (FHL124) possesses all four K(+)Cl(-) (KCC) cotransporter isoforms, KCC1-4, despite KCC2 being typically considered a neuronal isoform. Since at least two spliced variants, KCC2a and KCC2b, are co-expressed in cells of the central nervous system, this study sought to define the KCC2 expression profile in FHL124 cells. KCC2a, but not KCC2b transcripts were detected by reverse transcriptase polymerase chain reaction (RT-PCR). Proteins of molecular weights ranging from 95 to 135 kDa were found by Western blotting using non-variant specific anti-KCC2 antibodies directed against two different regions of the KCC2 proteins, and by biotinylation suggesting membrane expression. Immunofluorescence revealed membrane and punctate cytoplasmic staining for KCC2. Low levels of cytosolic αA and αB crystallines, and neuron-specific enolase were also detected contrasting with the strong membrane immunofluorescence staining for the Na/K ATPase α1 subunit. Since the lack of neuron-specific expression of the KCC2b variant in non-neuronal tissues has been proposed under control of a neuron-restrictive silencing element in the KCC2 gene, we hypothesize that this control may be lifted for the KCC2a variant in the FHL124 epithelial cell culture, a non-neuronal tissue of ectodermal origin.


Asunto(s)
Células Epiteliales/metabolismo , Simportadores/metabolismo , Secuencia de Bases , Línea Celular , Humanos , Inmunohistoquímica , Cristalino/citología , Microscopía Fluorescente , Datos de Secuencia Molecular , Mutación , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Isoformas de Proteínas/metabolismo , ARN Mensajero/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Simportadores/genética , Simportadores/inmunología , Cotransportadores de K Cl
17.
J Neurophysiol ; 108(3): 834-52, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22457464

RESUMEN

GABA depolarizes and excites central neurons during early development, becoming inhibitory and hyperpolarizing with maturation. This "developmental shift" occurs abruptly, reflecting a decrease in intracellular Cl(-) concentration ([Cl(-)](i)) and a hyperpolarizing shift in Cl(-) equilibrium potential due to upregulation of the K(+)-Cl(-) cotransporter KCC2b, a neuron-specific Cl(-) extruder. In contrast, primary afferent neurons (PANs) are depolarized by GABA throughout adulthood because of expression of NKCC1, a Na(+)-K(+)-2Cl(-) cotransporter that accumulates Cl(-) above equilibrium. The GABA(A)-mediated depolarization of PANs determines presynaptic inhibition in the spinal cord, a key mechanism gating somatosensory information. Little is known about developmental changes in Cl(-) transporter expression and Cl(-) homeostasis in PANs. Whether NKCC1 is expressed in PANs of all phenotypes or is restricted to subpopulations (e.g., nociceptors) is debatable. Likewise, whether PANs express KCC2s is controversial. We investigated NKCC1 and K(+)-Cl(-) cotransporter expression in rat and mouse dorsal root ganglion (DRG) neurons with molecular methods. Using fluorescence imaging microscopy, we measured [Cl(-)](i) in acutely dissociated rat DRG neurons (P0-P21) loaded with N-(ethoxycarbonylmethyl)-6-methoxyquinolinium bromide and classified with phenotypic markers. DRG neurons of all sizes express two NKCC1 mRNAs, one full-length and a shorter splice variant lacking exon 21. Immunolabeling with validated antibodies revealed ubiquitous expression of NKCC1 in DRG neurons irrespective of postnatal age and phenotype. As maturation progresses [Cl(-)](i) decreases gradually, persisting above equilibrium in >95% mature neurons. DRG neurons express mRNAs for KCC1, KCC3s, and KCC4, but not for KCC2s. Mechanisms underlying PANs' developmental changes in Cl(-) homeostasis are discussed and compared with those of central neurons.


Asunto(s)
Ganglios Espinales/crecimiento & desarrollo , Simportadores de Cloruro de Sodio-Potasio/fisiología , Simportadores/fisiología , Animales , Animales Recién Nacidos , Cloruros/análisis , Exones , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Compuestos de Quinolinio/farmacología , Ratas , Ratas Sprague-Dawley , Simportadores de Cloruro de Sodio-Potasio/biosíntesis , Simportadores de Cloruro de Sodio-Potasio/genética , Miembro 2 de la Familia de Transportadores de Soluto 12 , Simportadores/biosíntesis , Cotransportadores de K Cl
18.
J Biol Chem ; 287(1): 393-407, 2012 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-22094461

RESUMEN

Cell differentiation is compromised in acute leukemias. We report that mammalian target of rapamycin (mTOR) and S6 kinase (S6K) are highly expressed in the undifferentiated promyelomonocytic leukemic HL-60 cell line, whereas PLD2 expression is minimal. The expression ratio of PLD2 to mTOR (or to S6K) is gradually inverted upon in vitro induction of differentiation toward the neutrophilic phenotype. We present three ways that profoundly affect the kinetics of differentiation as follows: (i) simultaneous overexpression of mTOR (or S6K), (ii) silencing of mTOR via dsRNA-mediated interference or inhibition with rapamycin, and (iii) PLD2 overexpression. The last two methods shortened the time required for differentiation. By determining how PLD2 participates in cell differentiation, we found that PLD2 interacts with and activates the oncogene Fes/Fps, a protein-tyrosine kinase known to be involved in myeloid cell development. Fes activity is elevated with PLD2 overexpression, phosphatidic acid or phosphatidylinositol bisphosphate. Co-immunoprecipitation indicates a close PLD2-Fes physical interaction that is negated by a Fes-R483K mutant that incapacitates its Src homology 2 domain. All these suggest for the first time the following mechanism: mTOR/S6K down-regulation→PLD2 overexpression→PLD2/Fes association→phosphatidic acid-led activation of Fes kinase→granulocytic differentiation. Differentiation shortening could have a clinical impact on reducing the time of return to normalcy of the white cell counts after chemotherapy in patients with acute promyelocytic leukemia.


Asunto(s)
Diferenciación Celular , Leucemia Mieloide/patología , Fosfolipasa D/metabolismo , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Silenciador del Gen , Células HL-60 , Humanos , Cinética , Datos de Secuencia Molecular , Fosfolipasa D/genética , Proteínas Proto-Oncogénicas c-fes/química , Proteínas Proto-Oncogénicas c-fes/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Quinasas S6 Ribosómicas/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/deficiencia , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Dominios Homologos src
19.
J Biol Chem ; 285(25): 18991-9001, 2010 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-20410302

RESUMEN

The mammalian target of rapamycin (mTOR) and S6 kinase (S6K) pathway is essential for cell differentiation, growth, and survival. Phospholipase D2 (PLD2) plays a key role in mTOR/S6K mitogenic signaling. However, the impact of PLD on mTOR/S6K gene expression is not known. Here we show that interleukin-8 (IL-8) increases mRNA expression levels for PLD2, mTOR, and S6K, with PLD2 preceding mTOR/S6K in time. Silencing of PLD2 gene expression abrogated IL-8-induced mTOR/S6K mRNA expression, whereas silencing of mTOR or S6K gene expression resulted in large (>3-fold and >5-fold, respectively) increased levels of PLD2 RNA, which was paralleled by increases in protein expression and lipase activity. Treatment of cells with 0.5 nm rapamycin induced a similar trend. These results suggest that, under basal conditions, PLD2 expression and concomitant activity is negatively regulated by the mTOR/S6K signaling pathway. Down-regulation of PLD2 was confirmed in differentiated HL-60 leukocytes overexpressing an mTOR-wild type, but not an mTOR kinase-dead construct. At the cellular level, overexpression of mTOR-wild type resulted in lower basal cell migration, which was reversed by treatment with IL-8. We propose that IL-8 reverses an mTOR/S6K-led down-regulation of PLD2 expression and enables PLD2 to fully function as a facilitator for cell migration.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosfolipasa D/biosíntesis , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas S6 Ribosómicas/biosíntesis , Animales , Células COS , Diferenciación Celular , Chlorocebus aethiops , Silenciador del Gen , Células HL-60 , Humanos , Interleucina-8/metabolismo , Lipasa/metabolismo , Neutrófilos/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR
20.
Biochem Biophys Res Commun ; 389(2): 224-8, 2009 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-19715678

RESUMEN

Phospholipase D2 (PLD2) overexpression in mammalian cells results in cell transformation. We have hypothesized that this is due to an increase of de novo DNA synthesis. We show here that overexpression of PLD2-WT leads to an increased DNA synthesis, as measured by the expression levels of the proliferation markers PCNA, p27(KIP1) and phospho-histone-3. The enhancing effect was even higher with phosphorylation-deficient PLD2-Y179F and PLD2-Y511F mutants. The mechanism for this did not involve the enzymatic activity of the lipase, but, rather, the presence of the protein tyrosine phosphatase CD45, as silencing with siRNA for CD45 abrogated the effect. The two Y-->F mutants had in common a YxN consensus site that, in the phosphorylated counterparts, could be recognized by SH2-bearing proteins, such as Grb2. Even though Y179F and Y511F cannot bind Grb2, they could still find other protein partners, one of which, we have reasoned, could be CD45 itself. Affinity purified PLD2 is indeed activated by Grb2 and deactivated by CD45 in vitro. We concluded that phosphorylated PLD2, aided by Grb2, mediates lipase activity, whereas dephosphorylated PLD2 mediates an induction of cell proliferation, and the specific residues involved in this newly discovered regulation of PLD2 are Y(179) and Y(511).


Asunto(s)
Proliferación Celular , Transformación Celular Neoplásica/metabolismo , ADN/biosíntesis , Fosfolipasa D/metabolismo , Animales , Células COS , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Chlorocebus aethiops , Humanos , Datos de Secuencia Molecular , Fosfolipasa D/genética , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...