Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Arch Pharm (Weinheim) ; : e2400063, 2024 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-38704748

RESUMEN

Lithium induces nephrogenic diabetes insipidus (NDI) and microcystic chronic kidney disease (CKD). As previous clinical studies suggest that NDI is dose-dependent and CKD is time-dependent, we investigated the effect of low exposition to lithium in a long-term experimental rat model. Rats were fed with a normal diet (control group), with the addition of lithium (Li+ group), or with lithium and amiloride (Li+/Ami group) for 6 months, allowing obtaining low plasma lithium concentrations (0.25 ± 0.06 and 0.43 ± 0.16 mmol/L, respectively). Exposition to low concentrations of plasma lithium levels prevented NDI but not microcystic dilations of kidney tubules, which were identified as collecting ducts (CDs) on immunofluorescent staining. Both hypertrophy, characterized by an increase in the ratio of nuclei per tubular area, and microcystic dilations were observed. The ratio between principal cells and intercalated cells was higher in microcystic than in hypertrophied tubules. There was no correlation between AQP2 messenger RNA levels and cellular remodeling of the CD. Additional amiloride treatment in the Li+/Ami group did not allow consistent morphometric and cellular composition changes compared to the Li+ group. Low exposition to lithium prevented overt NDI but not microcystic dilations of the CD, with differential cellular composition in hypertrophied and microcystic CDs, suggesting different underlying cellular mechanisms.

2.
Int J Mol Sci ; 25(11)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38892145

RESUMEN

These last years, the growth factor GDF15 has emerged as a key element in many different biological processes. It has been established as being produced in response to many pathological states and is now referred to as a stress-related hormone. Regarding kidney functions, GDF15 has been involved in different pathologies such as chronic kidney disease, diabetic nephropathy, renal cancer, and so on. Interestingly, recent studies also revealed a role of GDF15 in the renal homeostatic mechanisms allowing to maintain constant, as far as possible, the plasma parameters such as pH and K+ values. In this review, we recapitulate the role of GDF15 in physiological and pathological context by focusing our interest on its renal effect.


Asunto(s)
Factor 15 de Diferenciación de Crecimiento , Riñón , Humanos , Factor 15 de Diferenciación de Crecimiento/metabolismo , Riñón/metabolismo , Riñón/fisiopatología , Animales , Enfermedades Renales/metabolismo , Enfermedades Renales/fisiopatología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/fisiopatología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/fisiopatología
3.
Int J Mol Sci ; 25(7)2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38612818

RESUMEN

Many genomic, anatomical and functional differences exist between the medullary (MTAL) and the cortical thick ascending limb of the loop of Henle (CTAL), including a higher expression of claudin-10 (CLDN10) in the MTAL than in the CTAL. Therefore, we assessed to what extent the Cldn10 gene expression is a determinant of differential gene expression between MTAL and CTAL. RNAs extracted from CTAL and MTAL microdissected from wild type (WT) and Cldn10 knock out mice (cKO) were analyzed by RNAseq. Differential and enrichment analyses (GSEA) were performed with interactive R Shiny software. Between WT and cKO MTAL, 637 genes were differentially expressed, whereas only 76 were differentially expressed between WT and cKO CTAL. Gene expression patterns and GSEA analyses in all replicates showed that WT MTAL did not cluster with the other replicates; no hierarchical clustering could be found between WT CTAL, cKO CTAL and cKO MTAL. Compared to WT replicates, cKO replicates were enriched in Cldn16, Cldn19, Pth1r, (parathyroid hormone receptor type 1), Casr (calcium sensing receptor) and Vdr (Vitamin D Receptor) mRNA in both the cortex and medulla. Cldn10 is associated with gene expression patterns, including genes specifically involved in divalent cations reabsorption in the TAL.


Asunto(s)
Médula Suprarrenal , Extremidades , Animales , Ratones , Claudinas/genética , Ratones Noqueados , Expresión Génica
4.
J Intern Med ; 293(1): 4-22, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35909256

RESUMEN

The kidneys, by equilibrating the outputs to the inputs, are essential for maintaining the constant volume, pH, and electrolyte composition of the internal milieu. Inability to do so, either because of internal kidney dysfunction (primary alteration) or because of some external factors (secondary alteration), leads to pathologies of varying severity, leading to modification of these parameters and affecting the functions of other organs. Alterations of the functions of the collecting duct (CD), the most distal part of the nephron, have been extensively studied and have led to a better diagnosis, better management of the related diseases, and the development of therapeutic tools. Thus, dysfunctions of principal cell-specific transporters such as ENaC or AQP2 or its receptors (mineralocorticoid or vasopressin receptors) caused by mutations or by compounds present in the environment (lithium, antibiotics, etc.) have been demonstrated in a variety of syndromes (Liddle, pseudohypoaldosteronism type-1, diabetes insipidus, etc.) affecting salt, potassium, and water balance. In parallel, studies on specific transporters (H+ -ATPase, anion exchanger 1) in intercalated cells have revealed the mechanisms of related tubulopathies like distal renal distal tubular acidosis or Sjögren syndrome. In this review, we will recapitulate the mechanisms of most of the primary and secondary alteration of the ion transport system of the CD to provide a better understanding of these diseases and highlight how a targeted perturbation may affect many different pathways due to the strong crosstalk and entanglements between the different actors (transporters, cell types).


Asunto(s)
Acidosis Tubular Renal , Túbulos Renales Colectores , Humanos , Túbulos Renales Colectores/metabolismo , Acuaporina 2/metabolismo , Nefronas/metabolismo , Riñón , Acidosis Tubular Renal/metabolismo , Agua/metabolismo
5.
J Am Soc Nephrol ; 31(5): 1009-1023, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32245797

RESUMEN

BACKGROUND: Water and solute transport across epithelia can occur via the transcellular or paracellular pathways. Tight junctions play a key role in mediating paracellular ion reabsorption in the kidney. In the renal collecting duct, which is a typical absorptive tight epithelium, coordination between transcellular sodium reabsorption and paracellular permeability may prevent the backflow of reabsorbed sodium to the tubular lumen along a steep electrochemical gradient. METHODS: To investigate whether transcellular sodium transport controls tight-junction composition and paracellular permeability via modulating expression of the transmembrane protein claudin-8, we used cultured mouse cortical collecting duct cells to see how overexpression or silencing of epithelial sodium channel (ENaC) subunits and claudin-8 affect paracellular permeability. We also used conditional kidney tubule-specific knockout mice lacking ENaC subunits to assess the ENaC's effect on claudin-8 expression. RESULTS: Overexpression or silencing of the ENaC γ-subunit was associated with parallel and specific changes in claudin-8 abundance. Increased claudin-8 abundance was associated with a reduction in paracellular permeability to sodium, whereas decreased claudin-8 abundance was associated with the opposite effect. Claudin-8 overexpression and silencing reproduced these functional effects on paracellular ion permeability. Conditional kidney tubule-specific ENaC γ-subunit knockout mice displayed decreased claudin-8 expression, confirming the cell culture experiments' findings. Importantly, ENaC ß-subunit or α-subunit silencing or kidney tubule-specific ß-ENaC or α-ENaC knockout mice did not alter claudin-8 abundance. CONCLUSIONS: Our data reveal the specific coupling between ENaC γ-subunit and claudin-8 expression. This coupling may play an important role in preventing the backflow of reabsorbed solutes and water to the tubular lumen, as well as in coupling paracellular and transcellular sodium permeability.


Asunto(s)
Claudinas/metabolismo , Canales Epiteliales de Sodio/metabolismo , Regulación de la Expresión Génica , Túbulos Renales Colectores/metabolismo , Sodio/metabolismo , Amilorida/análogos & derivados , Amilorida/farmacología , Animales , Transporte Biológico , Células Cultivadas , Cloruros/metabolismo , Claudinas/deficiencia , Claudinas/genética , Canales Epiteliales de Sodio/deficiencia , Canales Epiteliales de Sodio/genética , Silenciador del Gen , Transporte Iónico , Ratones , Ratones Noqueados , ARN Mensajero/biosíntesis , Proteínas Recombinantes/metabolismo , Transducción Genética
6.
Am J Physiol Renal Physiol ; 318(2): F422-F442, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31841389

RESUMEN

The thick ascending limb of the loop of Henle (TAL) is the first segment of the distal nephron, extending through the whole outer medulla and cortex, two regions with different composition of the peritubular environment. The TAL plays a critical role in the control of NaCl, water, acid, and divalent cation homeostasis, as illustrated by the consequences of the various monogenic diseases that affect the TAL. It delivers tubular fluid to the distal convoluted tubule and thereby affects the function of the downstream tubular segments. The TAL is commonly considered as a whole. However, many structural and functional differences exist between its medullary and cortical parts. The present review summarizes the available data regarding the similarities and differences between the medullary and cortical parts of the TAL. Both subsegments reabsorb NaCl and have high Na+-K+-ATPase activity and negligible water permeability; however, they express distinct isoforms of the Na+-K+-2Cl- cotransporter at the apical membrane. Ammonia and bicarbonate are mostly reabsorbed in the medullary TAL, whereas Ca2+ and Mg2+ are mostly reabsorbed in the cortical TAL. The peptidic hormone receptors controlling transport in the TAL are not homogeneously expressed along the cortical and medullary TAL. Besides this axial heterogeneity, structural and functional differences are also apparent between species, which underscores the link between properties and role of the TAL under various environments.


Asunto(s)
Corteza Renal/metabolismo , Médula Renal/metabolismo , Asa de la Nefrona/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Reabsorción Renal , Equilibrio Hidroelectrolítico , Adaptación Fisiológica , Animales , Evolución Molecular , Humanos , Corteza Renal/anatomía & histología , Médula Renal/anatomía & histología , Asa de la Nefrona/anatomía & histología , Proteínas de Transporte de Membrana/genética , Especificidad de la Especie
7.
Am J Physiol Regul Integr Comp Physiol ; 318(2): R320-R328, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31913688

RESUMEN

The modifications of the hemodynamic system and hydromineral metabolism are physiological features characterizing a normal gestation. Thus, the ability to expand plasma volume without increasing the level of blood pressure is necessary for the correct perfusion of the placenta. The kidney is essential in this adaptation by reabsorbing avidly sodium and fluid. In this study, we observed that the H,K-ATPase type 2 (HKA2), an ion pump expressed in kidney and colon and already involved in the control of the K+ balance during gestation, is also required for the correct plasma volume expansion and to maintain normal blood pressure. Indeed, compared with WT pregnant mice that exhibit a 1.6-fold increase of their plasma volume, pregnant HKA2-null mice (HKA2KO) only modestly expand their extracellular volume (×1.2). The renal expression of the epithelial Na channel (ENaC) α- and γ-subunits and that of the pendrin are stimulated in gravid WT mice, whereas the Na/Cl- cotransporter (NCC) expression is downregulated. These modifications are all blunted in HKA2KO mice. This impeded renal adaptation to gestation is accompanied by the development of hypotension in the pregnant HKA2KO mice. Altogether, our results showed that the absence of the HKA2 during gestation leads to an "underfilled" situation and has established this transporter as a key player of the renal control of salt and potassium metabolism during gestation.


Asunto(s)
Presión Sanguínea , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Riñón/enzimología , Volumen Plasmático , Potasio/metabolismo , Sodio/metabolismo , Animales , Acuaporina 2/metabolismo , Colon/enzimología , Canales Epiteliales de Sodio/genética , Canales Epiteliales de Sodio/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Edad Gestacional , ATPasa Intercambiadora de Hidrógeno-Potásio/deficiencia , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Homeostasis , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Transportadores de Sulfato/genética , Transportadores de Sulfato/metabolismo
8.
Nephrol Dial Transplant ; 35(11): 1901-1908, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31369102

RESUMEN

BACKGROUND: In rodents, the stimulation of adrenal progesterone is necessary for renal adaptation under potassium depletion. Here, we sought to determine the role of progesterone in adrenal adaptation in potassium-depleted healthy human volunteers and compared our findings with data collected in patients with Gitelman syndrome (GS), a salt-losing tubulopathy. METHODS: Twelve healthy young men were given a potassium-depleted diet for 7 days at a tertiary referral medical centre (NCT02297048). We measured by liquid chromatography coupled to tandem mass spectroscopy plasma steroid concentrations at Days 0 and 7 before and 30 min after treatment with tetracosactide. We compared these data with data collected in 10 GS patients submitted to tetracosactide test. RESULTS: The potassium-depleted diet decreased plasma potassium in healthy subjects by 0.3 ± 0.1 mmol/L, decreased plasma aldosterone concentration by 50% (P = 0.0332) and increased plasma 17-hydroxypregnenolone concentration by 45% (P = 0.0232) without affecting other steroids. CYP17 activity, as assessed by 17-hydroxypregnenolone/pregnenolone ratio, increased by 60% (P = 0.0389). As compared with healthy subjects, GS patients had 3-fold higher plasma concentrations of aldosterone, 11-deoxycortisol (+30%) and delta 4-androstenedione (+14%). Their post-tetracosactide progesterone concentration was 2-fold higher than that of healthy subjects and better correlated to plasma potassium than to plasma renin. CONCLUSION: The increase in 17-hydroxypregnenolone concentration after mild potassium depletion in otherwise healthy human subjects suggests that 17 hydroxylation of pregnenolone prevents the increase in progesterone observed in potassium-depleted mice. The unexpected over-response of non-mineralocorticoid steroids to tetracosactide in GS subjects suggests that the adrenal system not only adapts to sodium depletion but may also respond to hypokalaemia.


Asunto(s)
Glándulas Suprarrenales/fisiología , Síndrome de Gitelman/fisiopatología , Potasio/metabolismo , Progesterona/sangre , Adolescente , Adulto , Anciano , Aldosterona/sangre , Animales , Estudios de Casos y Controles , Cromatografía Liquida/métodos , Femenino , Síndrome de Gitelman/sangre , Humanos , Masculino , Ratones , Persona de Mediana Edad , Renina/sangre , Esteroides/sangre , Espectrometría de Masas en Tándem/métodos , Adulto Joven
9.
Am J Physiol Renal Physiol ; 317(2): F435-F443, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31188029

RESUMEN

We have recently reported that type A intercalated cells of the collecting duct secrete Na+ by a mechanism coupling the basolateral type 1 Na+-K+-2Cl- cotransporter with apical type 2 H+-K+-ATPase (HKA2) functioning under its Na+/K+ exchange mode. The first aim of the present study was to evaluate whether this secretory pathway is a target of atrial natriuretic peptide (ANP). Despite hyperaldosteronemia, metabolic acidosis is not associated with Na+ retention. The second aim of the present study was to evaluate whether ANP-induced stimulation of Na+ secretion by type A intercalated cells might account for mineralocorticoid escape during metabolic acidosis. In Xenopus oocytes expressing HKA2, cGMP, the second messenger of ANP, increased the membrane expression, activity, and Na+-transporting rate of HKA2. Feeding mice with a NH4Cl-enriched diet increased urinary excretion of aldosterone and induced a transient Na+ retention that reversed within 3 days. At that time, expression of ANP mRNA in the collecting duct and urinary excretion of cGMP were increased. Reversion of Na+ retention was prevented by treatment with an inhibitor of ANP receptors and was absent in HKA2-null mice. In conclusion, paracrine stimulation of HKA2 by ANP is responsible for the escape of the Na+-retaining effect of aldosterone during metabolic acidosis.


Asunto(s)
Equilibrio Ácido-Base , Acidosis/enzimología , Factor Natriurético Atrial/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Túbulos Renales Colectores/enzimología , Sodio/orina , Acidosis/genética , Acidosis/fisiopatología , Acidosis/orina , Adaptación Fisiológica , Aldosterona/orina , Animales , GMP Cíclico/orina , Femenino , ATPasa Intercambiadora de Hidrógeno-Potásio/deficiencia , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Concentración de Iones de Hidrógeno , Ratones Endogámicos C57BL , Ratones Noqueados , Comunicación Paracrina , Ratas , Transducción de Señal , Xenopus laevis
10.
Am J Physiol Renal Physiol ; 313(6): F1254-F1263, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28877883

RESUMEN

The cortical collecting duct (CCD) forms part of the aldosterone-sensitive distal nephron and plays an essential role in maintaining the NaCl balance and acid-base status. The CCD epithelium comprises principal cells as well as different types of intercalated cells. Until recently, transcellular Na+ transport was thought to be restricted to principal cells, whereas (acid-secreting) type A and (bicarbonate-secreting) type B intercalated cells were associated with the regulation of acid-base homeostasis. This review describes how this traditional view has been upended by several discoveries in the past decade. A series of studies has shown that type B intercalated cells can mediate electroneutral NaCl reabsorption by a mechanism involving Na+-dependent and Na+-independent Cl-/[Formula: see text] exchange, and that is energetically driven by basolateral vacuolar H+-ATPase pumps. Other research indicates that type A intercalated cells can mediate NaCl secretion, through a bumetanide-sensitive pathway that is energized by apical H+,K+-ATPase type 2 pumps operating as Na+/K+ exchangers. We also review recent findings on the contribution of the paracellular route to NaCl transport in the CCD. Last, we describe cross-talk processes, by which one CCD cell type impacts Na+/Cl- transport in another cell type. The mechanisms that have been identified to date demonstrate clearly the interdependence of NaCl and acid-base transport systems in the CCD. They also highlight the remarkable versatility of this nephron segment.


Asunto(s)
Transporte Biológico/fisiología , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Túbulos Renales Colectores/metabolismo , Nefronas/metabolismo , Cloruro de Sodio/metabolismo , Animales , Humanos , Sodio/metabolismo
11.
J Physiol ; 594(20): 5991-6008, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27412964

RESUMEN

KEY POINTS: The cortical collecting duct (CCD) plays an essential role in sodium homeostasis by fine-tuning the amount of sodium that is excreted in the urine. Ex vivo, the microperfused CCD reabsorbs sodium in the absence of lumen-to-bath concentration gradients. In the present study, we show that, in the presence of physiological lumen-to-bath concentration gradients, and in the absence of endocrine, paracrine and neural regulation, the mouse CCD secretes sodium, which represents a paradigm shift. This secretion occurs via the paracellular route, as well as a transcellular pathway that is energized by apical H+ /K+ -ATPase type 2 pumps operating as Na+ /K+ exchangers. The newly identified transcellular secretory pathway represents a physiological target for the regulation of sodium handling and for anti-hypertensive therapeutic agents. ABSTRACT: In vitro microperfusion experiments have demonstrated that cortical collecting ducts (CCDs) reabsorb sodium via principal and type B intercalated cells under sodium-depleted conditions and thereby contribute to sodium and blood pressure homeostasis. However, these experiments were performed in the absence of the transepithelial ion concentration gradients that prevail in vivo and determine paracellular transport. The present study aimed to characterize Na+ , K+ and Cl- fluxes in the mouse CCD in the presence of physiological transepithelial concentration gradients. For this purpose, we combined in vitro measurements of ion fluxes across microperfused CCDs of sodium-depleted mice with the predictions of a mathematical model. When NaCl transport was inhibited in all cells, CCDs secreted Na+ and reabsorbed K+ ; Cl- transport was negligible. Removing inhibitors of type A and B intercalated cells increased Na+ secretion in wild-type (WT) mice but not in H+ /K+ -ATPase type 2 (HKA2) knockout mice. Further inhibition of basolateral NaCl entry via the Na+ -K+ -2Cl- cotransporter in type A intercalated cells reduced Na+ secretion in WT mice to the levels observed in HKA2-/- mice. With no inhibitors, WT mouse CCDs still secreted Na+ and reabsorbed K+ . In vivo, HKA2-/- mice excreted less Na+ than WT mice after switching to a high-salt diet. Taken together, our results indicate that type A intercalated cells secrete Na+ via basolateral Na+ -K+ -2Cl- cotransporters in tandem with apical HKA2 pumps. They also suggest that the CCD can mediate overall Na+ secretion, and that its ability to reabsorb NaCl in vivo depends on the presence of acute regulatory factors.


Asunto(s)
Epitelio/metabolismo , Túbulos Renales Colectores/metabolismo , Animales , Transporte Biológico/fisiología , Cloruros/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Potasio/metabolismo , Sodio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
12.
Pflugers Arch ; 468(10): 1673-83, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27562425

RESUMEN

In industrialized countries, a large part of the population is daily exposed to low K(+) intake, a situation correlated with the development of salt-sensitive hypertension. Among many processes, adaptation to K(+)-restriction involves the stimulation of H,K-ATPase type 2 (HKA2) in the kidney and colon and, in this study, we have investigated whether HKA2 also contributes to the determination of blood pressure (BP). By using wild-type (WT) and HKA2-null mice (HKA2 KO), we showed that after 4 days of K(+) restriction, WT remain normokalemic and normotensive (112 ± 3 mmHg) whereas HKA2 KO mice exhibit hypokalemia and hypotension (104 ± 2 mmHg). The decrease of BP in HKA2 KO is due to the absence of NaCl-cotransporter (NCC) stimulation, leading to renal loss of salt and decreased extracellular volume (by 20 %). These effects are likely related to the renal resistance to vasopressin observed in HKA2 KO that may be explained, in part by the increased production of prostaglandin E2 (PGE2). In WT, the stimulation of NCC induced by K(+)-restriction is responsible for the elevation in BP when salt intake increases, an effect blunted in HKA2-null mice. The presence of an activated HKA2 is therefore required to limit the decrease in plasma [K(+)] but also contributes to the development of salt-sensitive hypertension.


Asunto(s)
ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Hipertensión/metabolismo , Deficiencia de Potasio/metabolismo , Potasio/metabolismo , Animales , Presión Sanguínea , Dinoprostona/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Hipertensión/etiología , Riñón/metabolismo , Ratones , Ratones Endogámicos C57BL , Potasio/sangre , Deficiencia de Potasio/complicaciones , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Vasopresinas/sangre
13.
Am J Physiol Renal Physiol ; 311(2): F469-86, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27194722

RESUMEN

It is now recognized that the metabolic disorders observed in diabetes are not, or not only due to the lack of insulin or insulin resistance, but also to elevated glucagon secretion. Accordingly, selective glucagon receptor antagonists are now proposed as a novel strategy for the treatment of diabetes. However, besides its metabolic actions, glucagon also influences kidney function. The glucagon receptor is expressed in the thick ascending limb, distal tubule, and collecting duct, and glucagon regulates the transepithelial transport of several solutes in these nephron segments. Moreover, it also influences solute transport in the proximal tubule, possibly by an indirect mechanism. This review summarizes the knowledge accumulated over the last 30 years about the influence of glucagon on the renal handling of electrolytes and urea. It also describes a possible novel role of glucagon in the short-term regulation of potassium homeostasis. Several original findings suggest that pancreatic α-cells may express a "potassium sensor" sensitive to changes in plasma K concentration and could respond by adapting glucagon secretion that, in turn, would regulate urinary K excretion. By their combined actions, glucagon and insulin, working in a combinatory mode, could ensure an independent regulation of both plasma glucose and plasma K concentrations. The results and hypotheses reviewed here suggest that the use of glucagon receptor antagonists for the treatment of diabetes should take into account their potential consequences on electrolyte handling by the kidney.


Asunto(s)
Glucagón/farmacología , Homeostasis/efectos de los fármacos , Riñón/efectos de los fármacos , Potasio/metabolismo , Animales , Humanos
14.
J Biol Chem ; 288(14): 10124-10131, 2013 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-23430254

RESUMEN

Proteinase-activated receptor 2 (PAR2) is a G protein-coupled membrane receptor that is activated upon cleavage of its extracellular N-terminal domain by trypsin and related proteases. PAR2 is expressed in kidney collecting ducts, a main site of control of Na(+) and K(+) homeostasis, but its function remains unknown. We evaluated whether and how PAR2 might control electrolyte transport in collecting ducts, and thereby participate in the regulation of blood pressure and plasma K(+) concentration. PAR2 is expressed at the basolateral border of principal and intercalated cells of the collecting duct where it inhibits K(+) secretion and stimulates Na(+) reabsorption, respectively. Invalidation of PAR2 gene impairs the ability of the kidney to control Na(+) and K(+) balance and promotes hypotension and hypokalemia in response to Na(+) and K(+) depletion, respectively. This study not only reveals a new role of proteases in the control of blood pressure and plasma potassium level, but it also identifies a second membrane receptor, after angiotensin 2 receptor, that differentially controls sodium reabsorption and potassium secretion in the late distal tubule. Conversely to angiotensin 2 receptor, PAR2 is involved in the regulation of sodium and potassium balance in the context of either stimulation or nonstimulation of the renin/angiotensin/aldosterone system. Therefore PAR2 appears not only as a new actor of the aldosterone paradox, but also as an aldosterone-independent modulator of blood pressure and plasma potassium.


Asunto(s)
Regulación de la Expresión Génica , Riñón/metabolismo , Potasio/sangre , Receptor PAR-2/metabolismo , Sodio/sangre , Aldosterona/metabolismo , Animales , Presión Sanguínea , Calcio/metabolismo , Diuréticos/farmacología , Homeostasis , Masculino , Ratones , Ratones Transgénicos , Perfusión , Ratas , Ratas Sprague-Dawley
15.
Am J Physiol Renal Physiol ; 306(7): F693-700, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24431203

RESUMEN

H-K-ATPase type 2 (HKA2), also known as the "nongastric" or "colonic" H-K-ATPase, is broadly expressed, and its presence in the kidney has puzzled experts in the field of renal ion transport systems for many years. One of the most important and robust characteristics of this transporter is that it is strongly stimulated after dietary K(+) restriction. This result prompted many investigators to propose that it should play a role in allowing the kidney to efficiently retain K(+) under K(+) depletion. However, the apparent absence of a clear renal phenotype in HKA2-null mice has led to the idea that this transporter is an epiphenomenon. This review summarizes past and recent findings regarding the functional, structural and physiological characteristics of H-K-ATPase type 2. The findings discussed in this review suggest that, as in the famous story, the ugly duckling of the X-K-ATPase family is actually a swan.


Asunto(s)
ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Riñón/enzimología , Animales , Transporte Biológico , Genotipo , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Homeostasis , Humanos , Isoenzimas , Fenotipo , Potasio/metabolismo
16.
Nephron ; : 1-27, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38824912

RESUMEN

BACKGROUND: Urea is the major end-product of protein metabolism in mammals. In carnivores and omnivores, a large load of urea is excreted daily in urine, with a concentration that is 30-100 times above that in plasma. This is important for the sake of water economy. Too little attention has been given to the existence of energy-dependent urea transport that plays an important role in this concentrating activity. SUMMARY: This review first presents functional evidence for an energy-dependent urea secretion that occurs exclusively in the straight part of the proximal tubule (PST). Second, it proposes a candidate transmembrane transporter responsible for this urea secretion in the PST. SLC6A18 is expressed exclusively in the PST and has been identified as a glycine transporter, based on findings in SLC6A18 knockout mice. We propose that it is actually a glycine/urea antiport, secreting urea into the lumen in exchange for glycine and Na. Glycine is most likely recycled back into the cell via a transporter located in the brush border. Urea secretion in the PST modifies the composition of the tubular fluid in the thick ascending limb and, thus, contributes, indirectly, to influence the "signal" at the macula densa that plays a crucial role in the regulation of the glomerular filtration rate (GFR) by the tubulo-glomerular feedback. KEY MESSAGES: Taking into account this secondary active secretion of urea in the mammalian kidney provides a new understanding of the influence of protein intake on GFR, of the regulation of urea excretion, and of the urine-concentrating mechanism.

17.
Function (Oxf) ; 5(4)2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38985001

RESUMEN

A detailed knowledge of the lipid composition of components of nephrons is crucial for understanding physiological processes and the development of kidney diseases. However, the lipidomic composition of kidney tubular segments is unknown. We manually isolated the proximal convoluted tubule (PCT), the cortical thick ascending limb of Henle's loop, and the cortical collecting duct from 5 lean and obese mice and subjected the samples to shotgun lipidomics analysis by high-resolution mass spectrometry acquisition. Across all samples, more than 500 lipid species were identified, quantified, and compared. We observed significant compositional differences among the 3 tubular segments, which serve as true signatures. These intrinsic lipidomic features are associated with a distinct proteomic program that regulates highly specific physiological functions. The distinctive lipidomic features of each of the 3 segments are mostly based on the relative composition of neutral lipids, long-chain polyunsaturated fatty acids, sphingolipids, and ether phospholipids. These features support the hypothesis of a lipotype assigned to specific tubular segments. Obesity profoundly impacts the lipotype of PCT. In conclusion, we present a comprehensive lipidomic analysis of 3 cortical segments of mouse kidney tubules. This valuable resource provides unparalleled detail that enhances our understanding of tubular physiology and the potential impact of pathological conditions.


Asunto(s)
Lipidómica , Animales , Ratones , Ratones Endogámicos C57BL , Masculino , Obesidad/metabolismo , Túbulos Renales Proximales/metabolismo , Corteza Renal/metabolismo , Corteza Renal/química , Lípidos/análisis , Metabolismo de los Lípidos/fisiología , Esfingolípidos/metabolismo
18.
iScience ; 27(6): 109737, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38799564

RESUMEN

Long-chain acyl-CoA synthetase family 4 (ACSL4) metabolizes long-chain polyunsaturated fatty acids (PUFAs), enriching cell membranes with phospholipids susceptible to peroxidation and drive ferroptosis. The role of ACSL4 and ferroptosis upon endoplasmic-reticulum (ER)-stress-induced acute kidney injury (AKI) is unknown. We used lipidomic, molecular, and cellular biology approaches along with a mouse model of AKI induced by ER stress to investigate the role of ACSL4 regulation in membrane lipidome remodeling in the injured tubular epithelium. Tubular epithelial cells (TECs) activate ACSL4 in response to STAT3 signaling. In this context, TEC membrane lipidome is remodeled toward PUFA-enriched triglycerides instead of PUFA-bearing phospholipids. TECs expressing ACSL4 in this setting are not vulnerable to ferroptosis. Thus, ACSL4 activity in TECs is driven by STAT3 signaling, but ACSL4 alone is not enough to sensitize ferroptosis, highlighting the significance of the biological context associated with the study model.

19.
Pflugers Arch ; 465(8): 1149-58, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23440459

RESUMEN

Renal K(+) retention is activated during pregnancy through a mechanism unknown to date. Here, we showed that the renal stimulation of H,K-ATPase type 2 (HKA2), whose expression was recently identified to be progesterone-dependent, is part of the mechanism favoring K(+) accumulation during gestation. Moreover, investigation of the gestational phenotype of HKA2-null mice compared to their wild-type (WT) littermate revealed a decrease in fertility (gestation was successful in 33 % of HKA2-null mice vs. 83 % of WT mice) and in litter size (6.5 ± 0.6 and 7.8 ± 0.4 fetuses per litter, respectively). We also observed that urinary K(+) excretion decreased by 20 % and plasma K(+) concentration rose slightly (11 %) in WT mice during gestation (relative to basal conditions). In contrast, the renal excretion of K(+) and plasma K(+) levels in HKA2-null mice remained constant during gestation, whereas fecal K(+) excretion increased. As a consequence, HKA2-null mice did not accumulate K(+) in their extracellular compartment as efficiently as WT mice did. Finally, the link between inefficient K(+) balance adaptations and gestational complications was established when we observed that these complications could be reversed with an increased K(+) uptake. Altogether, these results define a novel physiological role for the HKA2 transporter and uncover a link between K(+) metabolism and fertility.


Asunto(s)
Fertilidad/fisiología , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Homeostasis/fisiología , Riñón/metabolismo , Animales , Femenino , Riñón/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Potasio/metabolismo , Potasio/orina , Embarazo , Complicaciones del Embarazo/metabolismo , Complicaciones del Embarazo/fisiopatología
20.
FASEB J ; 26(7): 2859-67, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22459151

RESUMEN

Maintenance by the kidney of stable plasma K(+) values is crucial, as plasma K(+) controls muscle and nerve activity. Since renal K(+) excretion is regulated by the circadian clock, we aimed to identify the ion transporters involved in this process. In control mice, the renal mRNA expression of H,K-ATPase type 2 (HKA2) is 25% higher during rest compared to the activity period. Conversely, under dietary K(+) restriction, HKA2 expression is ∼40% higher during the activity period. This reversal suggests that HKA2 contributes to the circadian regulation of K(+) homeostasis. Compared to their wild-type (WT) littermates, HKA2-null mice fed a normal diet have 2-fold higher K(+) renal excretion during rest. Under K(+) restriction, their urinary K(+) loss is 40% higher during the activity period. This inability to excrete K(+) "on time" is reflected in plasma K(+) values, which vary by 12% between activity and rest periods in HKA2-null mice but remain stable in WT mice. Analysis of the circadian expression of HKA2 regulators suggests that Nrf2, but not progesterone, contributes to its rhythmicity. Therefore, HKA2 acts to maintain the circadian rhythm of urinary K(+) excretion and preserve stable plasma K(+) values throughout the day.


Asunto(s)
Ritmo Circadiano/fisiología , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Potasio/sangre , Animales , Proteínas CLOCK/deficiencia , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Ritmo Circadiano/genética , Regulación Enzimológica de la Expresión Génica , ATPasa Intercambiadora de Hidrógeno-Potásio/clasificación , ATPasa Intercambiadora de Hidrógeno-Potásio/deficiencia , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Homeostasis , Riñón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/metabolismo , Potasio/orina , Potasio en la Dieta/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA