Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 686
Filtrar
1.
Cell Stem Cell ; 29(11): 1510-1512, 2022 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-36332567

RESUMO

Collecting ducts in the kidney control the balance of water and electrolytes, as well as pH of the urine to maintain body homeostasis. Shi et al.1 recently reported in Nature Biotechnology a protocol to generate functional collecting duct cells from human pluripotent stem cells.


Assuntos
Túbulos Renais Coletores , Humanos , Túbulos Renais Coletores/fisiologia , Rim , Homeostase
2.
J Am Soc Nephrol ; 32(6): 1339-1354, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33727367

RESUMO

BACKGROUND: MicroRNAs (miRNAs), formed by cleavage of pre-microRNA by the endoribonuclease Dicer, are critical modulators of cell function by post-transcriptionally regulating gene expression. METHODS: Selective ablation of Dicer in AQP2-expressing cells (DicerAQP2Cre+ mice) was used to investigate the role of miRNAs in the kidney collecting duct of mice. RESULTS: The mice had severe polyuria and nephrogenic diabetes insipidus, potentially due to greatly reduced AQP2 and AQP4 levels. Although epithelial sodium channel levels were decreased in cortex and increased in inner medulla, amiloride-sensitive sodium reabsorption was equivalent in DicerAQP2Cre+ mice and controls. Small-RNA sequencing and proteomic analysis revealed 31 and 178 significantly regulated miRNAs and proteins, respectively. Integrated bioinformatic analysis of the miRNAome and proteome suggested alterations in the epigenetic machinery and various transcription factors regulating AQP2 expression in DicerAQP2Cre+ mice. The expression profile and function of three miRNAs (miR-7688-5p, miR-8114, and miR-409-3p) whose predicted targets were involved in epigenetic control (Phf2, Kdm5c, and Kdm4a) or transcriptional regulation (GATA3, GATA2, and ELF3) of AQP2 were validated. Luciferase assays could not demonstrate direct interaction of AQP2 or the three potential transcription factors with miR-7688-5p, miR-8114, and miR-409-3p. However, transfection of respective miRNA mimics reduced AQP2 expression. Chromatin immunoprecipitation assays demonstrated decreased Phf2 and significantly increased Kdm5c interactions at the Aqp2 gene promoter in DicerAQP2Cre+ mice, resulting in decreased RNA Pol II association. CONCLUSIONS: Novel evidence indicates miRNA-mediated epigenetic regulation of AQP2 expression.


Assuntos
Aquaporina 2/genética , Epigênese Genética/genética , Regulação da Expressão Gênica , MicroRNAs/genética , Ribonuclease III/genética , Animais , Aquaporina 2/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Diabetes Insípido Nefrogênico/genética , Diabetes Insípido Nefrogênico/metabolismo , Regulação para Baixo , Canais Epiteliais de Sódio/metabolismo , Feminino , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA3/genética , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Proteínas de Homeodomínio/genética , Túbulos Renais Coletores/fisiologia , Masculino , Camundongos , Poliúria/genética , Poliúria/metabolismo , Proteoma , Processamento Pós-Transcricional do RNA , Reabsorção Renal , Análise de Sequência de RNA , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
3.
Am J Physiol Cell Physiol ; 319(1): C136-C147, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32401606

RESUMO

The cortical collecting duct (CCD) of the mammalian kidney plays a major role in the maintenance of total body electrolyte, acid/base, and fluid homeostasis by tubular reabsorption and excretion. The mammalian CCD is heterogeneous, composed of Na+-absorbing principal cells (PCs) and acid-base-transporting intercalated cells (ICs). Perturbations in luminal flow rate alter hydrodynamic forces to which these cells in the cylindrical tubules are exposed. However, most studies of tubular ion transport have been performed in cell monolayers grown on or epithelial sheets affixed to a flat support, since analysis of transepithelial transport in native tubules by in vitro microperfusion requires considerable expertise. Here, we report on the generation and characterization of an in vitro, perfusable three-dimensional kidney CCD model (3D CCD), in which immortalized mouse PC-like mpkCCD cells are seeded within a cylindrical channel embedded within an engineered extracellular matrix and subjected to luminal fluid flow. We find that a tight epithelial barrier composed of differentiated and polarized PCs forms within 1 wk. Immunofluorescence microscopy reveals the apical epithelial Na+ channel ENaC and basolateral Na+/K+-ATPase. On cessation of luminal flow, benzamil-inhibitable cell doming is observed within these 3D CCDs consistent with the presence of ENaC-mediated Na+ absorption. Our 3D CCD provides a geometrically and microphysiologically relevant platform for studying the development and physiology of renal tubule segments.


Assuntos
Túbulos Renais Coletores/anatomia & histologia , Túbulos Renais Coletores/fisiologia , Modelos Biológicos , Perfusão/métodos , Impressão Tridimensional , Animais , Transporte Biológico/fisiologia , Linhagem Celular Transformada , Camundongos , Microscopia de Fluorescência/métodos
4.
Nephron ; 143(2): 148-152, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31408869

RESUMO

When injury occurs, it implies that attack has overcome defence. Tubulointerstitial injury plays important roles in acute kidney injury (AKI) and chronic kidney disease (CKD) and is the common pathway leading to end-stage renal disease, but how the renal tubulointerstitium defends against attack is poorly understood. Emerging evidence suggests that collecting ducts (CDs), which modify urine from nephrons and drain into ureter, could be key defenders protecting tubulointerstitium from injury; furthermore, the canonical renal vitamin A signalling physiologically confined to CDs could be a key regulator of this protective machinery. This hypothesis can be tested by in vitro, in vivo and clinical studies, particularly by repressing or boosting key molecular regulators in CDs, to observe the resulting phenotypes in models of AKI and CKD. Further investigation of this hypothesis could lead to new strategies for diagnosis, prevention and treatment of AKI and CKD.


Assuntos
Túbulos Renais Coletores/fisiologia , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/fisiopatologia , Animais , Progressão da Doença , Humanos , Modelos Biológicos , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/fisiopatologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
5.
Semin Nephrol ; 39(4): 353-367, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31300091

RESUMO

The epithelium of the kidney collecting duct (CD) is composed mainly of two different types of cells with distinct and complementary functions. CD principal cells traditionally have been considered to have a major role in Na+ and water regulation, while intercalated cells (ICs) were thought to largely modulate acid-base homeostasis. In recent years, our understanding of IC function has improved significantly owing to new research findings. Thus, we now have a new model for CD transport that integrates mechanisms of salt and water reabsorption, K+ homeostasis, and acid-base status between principal cells and ICs. There are three main types of ICs (type A, type B, and non-A, non-B), which first appear in the late distal convoluted tubule or in the connecting segment in a species-dependent manner. ICs can be detected in CD from cortex to the initial part of the inner medulla, although some transport proteins that are key components of ICs also are present in medullary CD, cells considered inner medullary. Of the three types of ICs, each has a distinct morphology and expresses different complements of membrane transport proteins that translate into very different functions in homeostasis and contributions to CD luminal pro-urine composition. This review includes recent discoveries in IC intracellular and paracrine signaling that contributes to acid-base regulation as well as Na+, Cl-, K+, and Ca2+ homeostasis. Thus, these new findings highlight the potential role of ICs as targets for potential hypertension treatments.


Assuntos
Equilíbrio Ácido-Base/fisiologia , Células Epiteliais/fisiologia , Túbulos Renais Coletores/fisiologia , Animais , Canais de Cálcio/fisiologia , Canais de Cloreto/fisiologia , Células Epiteliais/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Transporte de Íons/fisiologia , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/metabolismo , Canais de Potássio/fisiologia , Canais de Sódio/fisiologia
6.
G Ital Nefrol ; 35(6)2018 Dec.
Artigo em Italiano | MEDLINE | ID: mdl-30550035

RESUMO

ADH is a hormone secreted by neurohypophysis that plays different roles based on the target organ. At the renal level, this peptide is capable of causing electrolyte-free water absorption, thus playing a key role in the hydro-electrolytic balance. There are pathologies and disorders that jeopardize this balance and, in this field, ADH receptor inhibitors such as Vaptans could play a key role. By inhibiting the activation pathway of vasopressin, they are potentially useful in euvolemic and hypervolemic hypotonic hyponatremia. However, clinical trials in heart failure have not given favourable results on clinical outcomes. Even in SIADH, despite their wide use, there is no agreement by experts on their use. Since vaptans inhibit the cAMP pathway in tubular cells, their use has been proposed to inhibit cystogenesis. A clinical trial has shown favourable effects on ADPKD progression. Because vaptans have been shown to be effective in models of renal cysts disorders other than ADPKD, their use has been proposed in diseases such as nephronophthisis and recessive autosomal polycystic disease. Other possible uses of vaptans could be in kidney transplantation and cardiorenal syndrome. Due to the activity of ADH in coagulation and haemostasis, ADH's activation pathway by Desmopressin Acetate could be a useful strategy to reduce the risk of bleeding in biopsies in patients with haemorrhagic risk.


Assuntos
Antagonistas dos Receptores de Hormônios Antidiuréticos/uso terapêutico , Nefropatias/tratamento farmacológico , Terapia de Alvo Molecular , Neurofisinas/agonistas , Neurofisinas/antagonistas & inibidores , Precursores de Proteínas/agonistas , Precursores de Proteínas/antagonistas & inibidores , Receptores de Vasopressinas/efeitos dos fármacos , Vasopressinas/agonistas , Vasopressinas/antagonistas & inibidores , Desequilíbrio Hidroeletrolítico/tratamento farmacológico , Antagonistas dos Receptores de Hormônios Antidiuréticos/farmacologia , Cadáver , AMP Cíclico/fisiologia , Previsões , Humanos , Hiponatremia/tratamento farmacológico , Hiponatremia/fisiopatologia , Nefropatias/fisiopatologia , Doenças Renais Císticas/tratamento farmacológico , Transplante de Rim , Túbulos Renais Coletores/efeitos dos fármacos , Túbulos Renais Coletores/fisiologia , Neurofisinas/fisiologia , Rim Policístico Autossômico Dominante/tratamento farmacológico , Rim Policístico Autossômico Dominante/fisiopatologia , Precursores de Proteínas/fisiologia , Receptores de Vasopressinas/agonistas , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Doadores de Tecidos , Vasopressinas/fisiologia
7.
J Am Soc Nephrol ; 29(3): 857-868, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29237740

RESUMO

Collecting ducts make up the distal-most tubular segments of the kidney, extending from the cortex, where they connect to the nephron proper, into the medulla, where they release urine into the renal pelvis. During water deprivation, body water preservation is ensured by the selective transepithelial reabsorption of water into the hypertonic medullary interstitium mediated by collecting ducts. The collecting duct epithelium forms tight junctions composed of barrier-enforcing claudins and exhibits a higher transepithelial resistance than other segments of the renal tubule exhibit. However, the functional relevance of this strong collecting duct epithelial barrier is unresolved. Here, we report that collecting duct-specific deletion of an epithelial transcription factor, grainyhead-like 2 (GRHL2), in mice led to reduced expression of tight junction-associated barrier components, reduced collecting duct transepithelial resistance, and defective renal medullary accumulation of sodium and other osmolytes. In vitro, Grhl2-deficient collecting duct cells displayed increased paracellular flux of sodium, chloride, and urea. Consistent with these effects, Grhl2-deficient mice had diabetes insipidus, produced dilute urine, and failed to adequately concentrate their urine after water restriction, resulting in susceptibility to prerenal azotemia. These data indicate a direct functional link between collecting duct epithelial barrier characteristics, which appear to prevent leakage of interstitial osmolytes into urine, and body water homeostasis.


Assuntos
Epitélio/fisiologia , Túbulos Renais Coletores/fisiologia , Osmorregulação/genética , Junções Íntimas/genética , Junções Íntimas/fisiologia , Fatores de Transcrição/genética , Animais , Aquaporina 2/metabolismo , Aquaporina 4/metabolismo , Arginina Vasopressina/metabolismo , Azotemia/etiologia , Transporte Biológico/genética , Creatinina/urina , Perfilação da Expressão Gênica , Masculino , Camundongos , Concentração Osmolar , Transdução de Sinais , Ureia/metabolismo , Urina , Água/metabolismo , Privação de Água/fisiologia
8.
Dokl Biol Sci ; 473(1): 43-45, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28508198

RESUMO

In the course of adaptation of the rat kidney collecting duct cells to hypo-osmotic medium, the organic anion transporter inhibitor probenecid reduced significantly the regulatory cell volume decrease in response to a hypotonic shock. Both probenecid and hypotonic shock delayed significantly the entry into a cell of the fluorescent dye calcein, which exists as anion at neutral pH. Thus, the organic osmolyte transport plays an important role in the regulatory decrease of the principal cell volume under the hypo-osmotic conditions.


Assuntos
Túbulos Renais Coletores/fisiologia , Rim/fisiologia , Pressão Osmótica/fisiologia , Equilíbrio Hidroeletrolítico/fisiologia , Animais , Tamanho Celular/efeitos dos fármacos , Concentração de Íons de Hidrogênio , Rim/metabolismo , Túbulos Renais Coletores/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Probenecid/farmacologia , Ratos
9.
J Am Soc Nephrol ; 28(10): 2887-2900, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28507058

RESUMO

The transcription factor hepatocyte nuclear factor-1ß (HNF-1ß) is essential for normal kidney development and function. Inactivation of HNF-1ß in mouse kidney tubules leads to early-onset cyst formation and postnatal lethality. Here, we used Pkhd1/Cre mice to delete HNF-1ß specifically in renal collecting ducts (CDs). CD-specific HNF-1ß mutant mice survived long term and developed slowly progressive cystic kidney disease, renal fibrosis, and hydronephrosis. Compared with wild-type littermates, HNF-1ß mutant mice exhibited polyuria and polydipsia. Before the development of significant renal structural abnormalities, mutant mice exhibited low urine osmolality at baseline and after water restriction and administration of desmopressin. However, mutant and wild-type mice had similar plasma vasopressin and solute excretion levels. HNF-1ß mutant kidneys showed increased expression of aquaporin-2 mRNA but mislocalized expression of aquaporin-2 protein in the cytoplasm of CD cells. Mutant kidneys also had decreased expression of the UT-A urea transporter and collectrin, which is involved in apical membrane vesicle trafficking. Treatment of HNF-1ß mutant mIMCD3 cells with hypertonic NaCl inhibited the induction of osmoregulated genes, including Nr1h4, which encodes the transcription factor FXR that is required for maximal urinary concentration. Chromatin immunoprecipitation and sequencing experiments revealed HNF-1ß binding to the Nr1h4 promoter in wild-type kidneys, and immunoblot analysis revealed downregulated expression of FXR in HNF-1ß mutant kidneys. These findings reveal a novel role of HNF-1ß in osmoregulation and identify multiple mechanisms, whereby mutations of HNF-1ß produce defects in urinary concentration.


Assuntos
Fator 1-beta Nuclear de Hepatócito/fisiologia , Túbulos Renais Coletores/fisiologia , Animais , Linhagem Celular , Feminino , Regulação da Expressão Gênica , Masculino , Camundongos Transgênicos , Poliúria/genética , Regiões Promotoras Genéticas , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Urina
10.
Results Probl Cell Differ ; 60: 165-203, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28409346

RESUMO

The collecting duct of the mammalian kidney is important for the regulation of extracellular volume, osmolarity, and pH. There are two major structurally and functionally distinct cell types: principal cells and intercalated cells. The former regulates Na+ and water homeostasis, while the latter participates in acid-base homeostasis. In vivo lineage tracing using Cre recombinase or its derivatives such as CreGFP and CreERT2 is a powerful new technique to identify stem/progenitor cells in their native environment and to decipher the origins of the tissue that they give rise to. Recent studies using this technique in mice have revealed multiple renal progenitor cell populations that differentiate into various nephron segments and collecting duct. In particular, emerging evidence suggests that like principal cells, most of intercalated cells originate from the progenitor cells expressing water channel Aquaporin 2. Mutations or malfunctions of the channels, pumps, and transporters expressed in the collecting duct system cause various human diseases. For example, gain-of-function mutations in ENaC cause Liddle's syndrome, while loss-of-function mutations in ENaC lead to Pseudohypoaldosteronism type 1. Mutations in either AE1 or V-ATPase B1 result in distal renal tubular acidosis. Patients with disrupted AQP2 or AVPR2 develop nephrogenic diabetes insipidus. A better understanding of the function and development of the collecting duct system may facilitate the discovery of new therapeutic strategies for treating kidney disease.


Assuntos
Túbulos Renais Coletores/embriologia , Túbulos Renais Coletores/fisiologia , Organogênese/fisiologia , Células-Tronco/citologia , Animais , Humanos , Nefropatias/genética , Túbulos Renais Coletores/patologia
11.
Anat Rec (Hoboken) ; 299(7): 967-72, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27111677

RESUMO

The ovine kidney has been recently determined to be a better model than the swine kidney for the study of collecting system healing after partial nephrectomy. However, there is no histological study comparing the collecting systems of these species. To compare human, swine, and ovine collecting systems using histomorphometry. The collecting systems of 10 kidneys from each species (human, swine, and ovine) were processed for histomorphometry. The thickness of the three layers (mucosal connective tissue, submucosal muscular, and adventitial connective tissue) were measured. The densities of smooth muscle fibers, elastic system fibers, and cells were also measured. Additionally, blood vessel density in the adventitial connective tissue was measured. Analysis of the collecting systems from the three species presented several differences. The adventitial connective tissue from the swine samples was thicker, with more blood vessels and smooth muscle fibers, compared with that from the human and ovine samples. Swine also had higher density of elastic fibers on the submucosal muscular layer. Ovine and human collecting systems shared several similar features, such as blood vessel and elastic fiber density in all layers and the density of cellular and muscular fibers in the submucosal muscular and adventitial connective tissue layers. The collecting system of the ovine kidney is more similar to that of the human kidney compared with that of the swine kidney. This may explain the differences between the healing mechanisms in swine and those in humans and sheep after partial nephrectomy. Anat Rec, 299:967-972, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Túbulos Renais Coletores/anatomia & histologia , Músculo Liso/anatomia & histologia , Nefrectomia , Cicatrização , Animais , Humanos , Imuno-Histoquímica , Túbulos Renais Coletores/fisiologia , Músculo Liso/fisiologia , Ovinos , Suínos
12.
J Ethnopharmacol ; 185: 310-8, 2016 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-26979340

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Oryeongsan (ORS, Wulingsan) has been reported to possess renal protective effects from renal diseases such as diabetes-induced renal damage, and nephrocalcinosis. AIM OF THE STUDY: This study was conducted to evaluate the quantitative analysis and the inhibitory effect of ORS on hypertonic stress-induced water channel and apoptosis in murine inner medullary collecting duct cell line (mIMCD-3). MATERIALS AND METHODS: Chromatographic and NMR spectroscopic analysis were performed and water balance regulation was determined by Western blot, RT-PCR, and immunofluorescnece. RESULTS: Seven active principles (5-hydroxymethylfurfural, alismoxide, methyl(-)trans-cinnamate, adenine, guanosine, adenosine, and ferulic acid) in ORS were isolated and the structures were identified mainly by NMR spectroscopic analysis. In addition, contents of these metabolites in ORS were evaluated by HPLC analysis. Pretreatment with ORS significantly attenuated the hypertonic stress (175mM NaCl)-induced increase in protein levels of AQP2 and apical membrane insertion. ORS also attenuated osmolyte sodium-myo-inositol transporter (SMIT) expression and tonicity-responsive enhancer binding protein (TonEBP) mRNA under hypertonic stress. Those actions of ORS presented the similar effect of PKA inhibitor which AQP2 expression throughout the inhibition of vasopressin-mediated cAMP/PKA signal pathway. On the other hand, pretreatment with ORS attenuated hypertonic stress-induced cell death. Hypertonic stress-induced Bax or caspase-3 expression was decreased by ORS, resulting in anti-apoptotic effect. CONCLUSIONS: The present data suggest that the beneficial effect of ORS in water balance and apoptosis against hypertonic stress of renal collecting ducts.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/efeitos dos fármacos , Água/metabolismo , Animais , Aquaporina 2/genética , Aquaporina 2/metabolismo , Biotinilação , Linhagem Celular , Medicamentos de Ervas Chinesas/química , Regulação da Expressão Gênica/fisiologia , Túbulos Renais Coletores/fisiologia , Camundongos , Pressão Osmótica/efeitos dos fármacos
13.
Pflugers Arch ; 468(7): 1197-1206, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27023350

RESUMO

The collecting duct (CD) is the final segment of the kidney involved in the fine regulation of osmotic and ionic balance. During dehydration, arginine vasopressin (AVP) stimulates the expression and trafficking of aquaporin 2 (AQP2) to the apical membrane of CD principal cells, thereby allowing water reabsorption from the primary urine. Conversely, when the secretion of AVP is lowered, as for instance upon water ingestion or as a consequence of diabetes insipidus, the CD remains water impermeable leading to enhanced diuresis and urine dilution. In addition, an AVP-independent mechanism of urine dilution is also at play when fasting. Piezo1/2 are recently discovered essential components of the non-selective mechanically activated cationic channels. Using quantitative PCR analysis and taking advantage of a ß-galactosidase reporter mouse, we demonstrate that Piezo1 is preferentially expressed in CD principal cells of the inner medulla at the adult stage, unlike Piezo2. Remarkably, siRNAs knock-down or conditional genetic deletion of Piezo1 specifically in renal cells fully suppresses activity of the stretch-activated non-selective cationic channels (SACs). Piezo1 in CD cells is dispensable for urine concentration upon dehydration. However, urinary dilution and decrease in urea concentration following rehydration are both significantly delayed in the absence of Piezo1. Moreover, decreases in urine osmolarity and urea concentration associated with fasting are fully impaired upon Piezo1 deletion in CD cells. Altogether, these findings indicate that Piezo1 is critically required for SAC activity in CD principal cells and is implicated in urinary osmoregulation.


Assuntos
Canais Iônicos/metabolismo , Túbulos Renais Coletores/metabolismo , Túbulos Renais Coletores/fisiologia , Equilíbrio Hidroeletrolítico/fisiologia , Animais , Aquaporina 2/metabolismo , Arginina Vasopressina/farmacologia , Linhagem Celular , Desidratação/metabolismo , Desidratação/fisiopatologia , Diurese/fisiologia , Túbulos Renais Coletores/efeitos dos fármacos , Camundongos , Concentração Osmolar , Equilíbrio Hidroeletrolítico/efeitos dos fármacos
14.
PLoS One ; 10(8): e0134477, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26280830

RESUMO

The kidney is one of the main organs that produces ammonia and release it into the circulation. Under normal conditions, between 30 and 50% of the ammonia produced in the kidney is excreted in the urine, the rest being absorbed into the systemic circulation via the renal vein. In acidosis and in some pathological conditions, the proportion of urinary excretion can increase to 70% of the ammonia produced in the kidney. Mechanisms regulating the balance between urinary excretion and renal vein release are not fully understood. We developed a mathematical model that reflects current thinking about renal ammonia handling in order to investigate the role of each tubular segment and identify some of the components which might control this balance. The model treats the movements of water, sodium chloride, urea, NH3 and [Formula: see text], and non-reabsorbable solute in an idealized renal medulla of the rat at steady state. A parameter study was performed to identify the transport parameters and microenvironmental conditions that most affect the rate of urinary ammonia excretion. Our results suggest that urinary ammonia excretion is mainly determined by those parameters that affect ammonia recycling in the loops of Henle. In particular, our results suggest a critical role for interstitial pH in the outer medulla and for luminal pH along the inner medullary collecting ducts.


Assuntos
Amônia/urina , Medula Renal/fisiologia , Túbulos Renais Coletores/fisiologia , Alça do Néfron/fisiologia , Modelos Biológicos , Algoritmos , Compostos de Amônio/análise , Animais , Simulação por Computador , Concentração de Íons de Hidrogênio , Ratos , Cloreto de Sódio/análise , Ureia/análise , Água/análise
15.
Am J Physiol Renal Physiol ; 309(2): F154-63, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25972513

RESUMO

The present study explored whether the intercalated cell Cl(-)/HCO3(-) exchanger pendrin modulates epithelial Na(+) channel (ENaC) function by changing channel open probability and/or channel density. To do so, we measured ENaC subunit subcellular distribution by immunohistochemistry, single channel recordings in split open cortical collecting ducts (CCDs), as well as transepithelial voltage and Na(+) absorption in CCDs from aldosterone-treated wild-type and pendrin-null mice. Because pendrin gene ablation reduced 70-kDa more than 85-kDa γ-ENaC band density, we asked if pendrin gene ablation interferes with ENaC cleavage. We observed that ENaC-cleaving protease application (trypsin) increased the lumen-negative transepithelial voltage in pendrin-null mice but not in wild-type mice, which raised the possibility that pendrin gene ablation blunts ENaC cleavage, thereby reducing open probability. In mice harboring wild-type ENaC, pendrin gene ablation reduced ENaC-mediated Na(+) absorption by reducing channel open probability as well as by reducing channel density through changes in subunit total protein abundance and subcellular distribution. Further experiments used mice with blunted ENaC endocytosis and degradation (Liddle's syndrome) to explore the significance of pendrin-dependent changes in ENaC open probability. In mouse models of Liddle's syndrome, pendrin gene ablation did not change ENaC subunit total protein abundance, subcellular distribution, or channel density, but markedly reduced channel open probability. We conclude that in mice harboring wild-type ENaC, pendrin modulates ENaC function through changes in subunit abundance, subcellular distribution, and channel open probability. In a mouse model of Liddle's syndrome, however, pendrin gene ablation reduces channel activity mainly through changes in open probability.


Assuntos
Proteínas de Transporte de Ânions/fisiologia , Canais Epiteliais de Sódio/metabolismo , Túbulos Renais Coletores/fisiologia , Sódio/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Síndrome de Liddle/genética , Síndrome de Liddle/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transportadores de Sulfato , Tripsina
16.
Clin J Am Soc Nephrol ; 10(2): 305-24, 2015 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-25632105

RESUMO

Intercalated cells are kidney tubule epithelial cells with important roles in the regulation of acid-base homeostasis. However, in recent years the understanding of the function of the intercalated cell has become greatly enhanced and has shaped a new model for how the distal segments of the kidney tubule integrate salt and water reabsorption, potassium homeostasis, and acid-base status. These cells appear in the late distal convoluted tubule or in the connecting segment, depending on the species. They are most abundant in the collecting duct, where they can be detected all the way from the cortex to the initial part of the inner medulla. Intercalated cells are interspersed among the more numerous segment-specific principal cells. There are three types of intercalated cells, each having distinct structures and expressing different ensembles of transport proteins that translate into very different functions in the processing of the urine. This review includes recent findings on how intercalated cells regulate their intracellular milieu and contribute to acid-base regulation and sodium, chloride, and potassium homeostasis, thus highlighting their potential role as targets for the treatment of hypertension. Their novel regulation by paracrine signals in the collecting duct is also discussed. Finally, this article addresses their role as part of the innate immune system of the kidney tubule.


Assuntos
Equilíbrio Ácido-Base , Células Epiteliais/fisiologia , Túbulos Renais Coletores/fisiologia , Acidose Tubular Renal/metabolismo , Acidose Tubular Renal/fisiopatologia , Animais , Diferenciação Celular , Linhagem da Célula , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Humanos , Imunidade Inata , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/imunologia , Túbulos Renais Coletores/metabolismo , Fenótipo , Sistema Renina-Angiotensina
17.
Clin J Am Soc Nephrol ; 10(5): 852-62, 2015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-25078421

RESUMO

Alterations in water homeostasis can disturb cell size and function. Although most cells can internally regulate cell volume in response to osmolar stress, neurons are particularly at risk given a combination of complex cell function and space restriction within the calvarium. Thus, regulating water balance is fundamental to survival. Through specialized neuronal "osmoreceptors" that sense changes in plasma osmolality, vasopressin release and thirst are titrated in order to achieve water balance. Fine-tuning of water absorption occurs along the collecting duct, and depends on unique structural modifications of renal tubular epithelium that confer a wide range of water permeability. In this article, we review the mechanisms that ensure water homeostasis as well as the fundamentals of disorders of water balance.


Assuntos
Encéfalo/citologia , Homeostase/fisiologia , Medula Renal/fisiologia , Equilíbrio Hidroeletrolítico/fisiologia , Água/fisiologia , Tamanho Celular , Diabetes Insípido/fisiopatologia , Humanos , Hiponatremia/fisiopatologia , Túbulos Renais Coletores/fisiologia , Pressão Osmótica , Células Receptoras Sensoriais , Sede/fisiologia , Vasopressinas/fisiologia
18.
Cell Physiol Biochem ; 34(5): 1802-11, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25502637

RESUMO

BACKGROUND/AIMS: Renal principal cells maintain their intracellular water and electrolyte content despite significant fluctuations of the extracellular water and salt concentrations. Their water permeability decreases rapidly (within a few seconds) after successive hypo-osmotic shocks. Our aim was to investigate the contribution of the apical and basolateral surface to this effect and the potential influence of fast reduction in AQP-2, -3 or -4 plasma membrane content. METHODS: Rat principal cells of kidney collecting duct fragments underwent hypo-osmotic challenge applied apically or basolaterally and the regulatory volume decrease (RVD) was measured by the calcein quenching method. The AQP -2, -3 and -4 content of the plasma membrane fraction was quantified by Western blotting. RESULTS: The hypo-osmotic shock applied apically causes rapid swelling with high apparent water permeability and fast RVD. An identical successive shock after 15-20 sec causes significantly lower swelling rate with 3-fold reduction in apparent water permeability. This reaction is accompanied by AQP2 decrease in the plasma membrane while AQP3 and AQP4 are unaffected. The contribution of the basolateral cell surface to RVD is significantly lower than the apical. CONCLUSION: These results indicate that in principal cells the effective mechanism of RVD is mainly regulated by the apical cell plasma membrane.


Assuntos
Permeabilidade da Membrana Celular/fisiologia , Túbulos Renais Coletores/fisiologia , Osmose/fisiologia , Pressão Osmótica/fisiologia , Água/metabolismo , Animais , Aquaporinas/metabolismo , Membrana Celular/metabolismo , Membrana Celular/fisiologia , Túbulos Renais Coletores/metabolismo , Ratos , Ratos Wistar , Equilíbrio Hidroeletrolítico/fisiologia
19.
Curr Pediatr Rev ; 10(2): 107-14, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25088264

RESUMO

Renal development begins in-utero and continues throughout childhood. Almost one-third of all developmental anomalies include structural or functional abnormalities of the urinary tract. There are three main phases of in-utero renal development: Pronephros, Mesonephros and Metanephros. Within three weeks of gestation, paired pronephri appear. A series of tubules called nephrotomes fuse with the pronephric duct. The pronephros elongates and induces the nearby mesoderm, forming the mesonephric (Woffian) duct. The metanephros is the precursor of the mature kidney that originates from the ureteric bud and the metanephric mesoderm (blastema) by 5 weeks of gestation. The interaction between these two components is a reciprocal process, resulting in the formation of a mature kidney. The ureteric bud forms the major and minor calyces, and the collecting tubules while the metanephrogenic blastema develops into the renal tubules and glomeruli. In humans, all of the nephrons are formed by 32 to 36 weeks of gestation. Simultaneously, the lower urinary tract develops from the vesico urethral canal, ureteric bud and mesonephric duct. In utero, ureters deliver urine from the kidney to the bladder, thereby creating amniotic fluid. Transcription factors, extracellular matrix glycoproteins, signaling molecules and receptors are the key players in normal renal development. Many medications (e.g., aminoglycosides, cyclooxygenase inhibitors, substances that affect the renin-angiotensin aldosterone system) also impact renal development by altering the expression of growth factors, matrix regulators or receptors. Thus, tight regulation and coordinated processes are crucial for normal renal development.


Assuntos
Túbulos Renais Coletores/embriologia , Mesonefro/embriologia , Pronefro/embriologia , Sistema Renina-Angiotensina/fisiologia , Sistema Urinário/embriologia , Sistema Urogenital/embriologia , Diferenciação Celular , Matriz Extracelular , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Túbulos Renais Coletores/anatomia & histologia , Túbulos Renais Coletores/fisiologia , Mesonefro/anatomia & histologia , Mesonefro/fisiologia , Pronefro/anatomia & histologia , Pronefro/fisiologia , Transdução de Sinais , Sistema Urinário/anatomia & histologia , Sistema Urogenital/anatomia & histologia , Sistema Urogenital/fisiologia
20.
Am J Physiol Renal Physiol ; 307(5): F593-600, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24990896

RESUMO

The localization and regulation of (pro)renin receptor (PRR) expression in kidney collecting duct cells are not well established. We hypothesized that low salt (LS) contributes to the regulation of PRR expression in these cells via the GSK-3ß-NFAT5-sirtuin1 (SIRT-1) signaling pathway. Mouse inner medullary collecting duct (mIMCD) cells were treated with NaCl at 130 (normal salt; NS), 63 (LS), or 209 mM (high salt; HS) alone or in combination with NFAT5 scrambled small interfering (si) RNA, NFAT5 siRNA, or the SIRT-1 inhibitor EX-527. Compared with NS, LS increased the mRNA and protein expression of PRR by 71% and 69% (P < 0.05), and reduced phosphorylation of GSK-3ß by 62% (P < 0.01), mRNA and protein expressions of NFAT5 by 65% and 45% (P < 0.05), and SIRT-1 by 44% and 50% (P < 0.01), respectively. LS also enhanced p65 NF-κB by 102% (P < 0.01). Treatment with HS significantly reduced the mRNA and protein expression of PRR by 32% and 23% (P < 0.05), and increased the mRNA and protein expression of NFAT5 by 39% and 45% (P < 0.05) and SIRT-1 by 51% and 56% (P < 0.05), respectively. HS+NFAT5 siRNA reduced the mRNA and protein expression of NFAT5 by 51% and 35% (P < 0.01) and increased the mRNA and protein expression of PRR by 148% and 70% (P < 0.01), respectively. HS+EX-527 significantly increased the mRNA and protein expression of PRR by 96% and 58% (P < 0.05), respectively. We conclude that expression of PRR in mIMCD cells is regulated by the GSK-3ß-NFAT5- SIRT-1 signaling pathway.


Assuntos
Quinase 3 da Glicogênio Sintase/fisiologia , Túbulos Renais Coletores/fisiologia , Receptores de Superfície Celular/fisiologia , Transdução de Sinais/fisiologia , Sirtuína 1/fisiologia , Fatores de Transcrição/fisiologia , Animais , Carbazóis/farmacologia , Linhagem Celular , Relação Dose-Resposta a Droga , Quinase 3 da Glicogênio Sintase/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta , Técnicas In Vitro , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/efeitos dos fármacos , Camundongos , Fosforilação , RNA Interferente Pequeno/farmacologia , Receptores de Superfície Celular/efeitos dos fármacos , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/efeitos dos fármacos , Cloreto de Sódio/farmacologia , Fator de Transcrição RelA/fisiologia , Fatores de Transcrição/efeitos dos fármacos , Receptor de Pró-Renina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA