Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Pflugers Arch ; 476(4): 555-564, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38195948

RESUMO

The kidney plays a crucial role in acid-base homeostasis. In the distal nephron, α-intercalated cells contribute to urinary acid (H+) secretion and ß-intercalated cells accomplish urinary base (HCO3-) secretion. ß-intercalated cells regulate the acid base status through modulation of the apical Cl-/HCO3- exchanger pendrin (SLC26A4) activity. In this review, we summarize and discuss our current knowledge of the physiological role of the renal transporter AE4 (SLC4A9). The AE4, as cation-dependent Cl-/HCO3- exchanger, is exclusively expressed in the basolateral membrane of ß-intercalated cells and is essential for the sensing of metabolic acid-base disturbances in mice, but not for renal sodium reabsorption and plasma volume control. Potential intracellular signaling pathways are discussed that might link basolateral acid-base sensing through the AE4 to apical pendrin activity.


Assuntos
Túbulos Renais Coletores , Animais , Camundongos , Antiportadores de Cloreto-Bicarbonato/metabolismo , Rim/metabolismo , Túbulos Renais Coletores/metabolismo
2.
Pflugers Arch ; 476(4): 545-554, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38221598

RESUMO

Secretin is a key hormone of the intestinal phase of digestion which activates pancreatic, bile duct and Brunner gland HCO3- secretion. Recently, the secretin receptor (SCTR) was also found in the basolateral membrane of the beta-intercalated cell (B-IC) of the collecting duct. Experimental addition of secretin triggers a pronounced activation of urinary HCO3- excretion, which is fully dependent on key functional proteins of the B-IC, namely apical pendrin and CFTR and the basolateral SCTR. Recent studies demonstrated that the SCTR knock-out mouse is unable to respond to an acute base load. Here, SCTR KO mice could not rapidly increase urine base excretion, developed prolonged metabolic alkalosis and exhibited marked compensatory hypoventilation. Here, we review the physiological effects of secretin with distinct focus on how secretin activates renal HCO3- excretion. We describe its new function as a hormone for HCO3- homeostasis.


Assuntos
Bicarbonatos , Secretina , Camundongos , Animais , Secretina/metabolismo , Secretina/farmacologia , Membrana Celular/metabolismo , Transportadores de Sulfato/metabolismo , Transporte Biológico , Homeostase , Bicarbonatos/metabolismo
3.
Am J Physiol Renal Physiol ; 324(1): F12-F29, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36264886

RESUMO

The renal response to acid-base disturbances involves phenotypic and remodeling changes in the collecting duct. This study examines whether the proximal tubule controls these responses. We examined mice with genetic deletion of proteins present only in the proximal tubule, either the A variant or both A and B variants of isoform 1 of the electrogenic Na+-bicarbonate cotransporter (NBCe1). Both knockout (KO) mice have spontaneous metabolic acidosis. We then determined the collecting duct phenotypic responses to this acidosis and the remodeling responses to exogenous acid loading. Despite the spontaneous acidosis in NBCe1-A KO mice, type A intercalated cells in the inner stripe of the outer medullary collecting duct (OMCDis) exhibited decreased height and reduced expression of H+-ATPase, anion exchanger 1, Rhesus B glycoprotein, and Rhesus C glycoprotein. Combined kidney-specific NBCe1-A/B deletion induced similar changes. Ultrastructural imaging showed decreased apical plasma membrane and increased vesicular H+-ATPase in OMCDis type A intercalated cell in NBCe1-A KO mice. Next, we examined the collecting duct remodeling response to acidosis. In wild-type mice, acid loading increased the proportion of type A intercalated cells in the connecting tubule (CNT) and OMCDis, and it decreased the proportion of non-A, non-B intercalated cells in the connecting tubule, and type B intercalated cells in the cortical collecting duct (CCD). These changes were absent in NBCe1-A KO mice. We conclude that the collecting duct phenotypic and remodeling responses depend on proximal tubule-dependent signaling mechanisms blocked by constitutive deletion of proximal tubule NBCe1 proteins.NEW & NOTEWORTHY This study shows that the proximal tubule regulates collecting duct phenotypic and remodeling responses to acidosis.


Assuntos
Acidose , Túbulos Renais Coletores , Simportadores de Sódio-Bicarbonato , Animais , Camundongos , Acidose/genética , Acidose/metabolismo , Glicoproteínas/metabolismo , Túbulos Renais Coletores/metabolismo , Túbulos Renais Proximais/metabolismo , Camundongos Knockout , ATPases Translocadoras de Prótons/metabolismo , Simportadores de Sódio-Bicarbonato/metabolismo
4.
Physiol Rep ; 10(14): e15362, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35851763

RESUMO

SLC26A4/Pendrin is the major electroneutral Cl- /HCO3- exchanger of the apical membrane of the Type B intercalated cell (IC) of the connecting segment (CNT) and cortical collecting duct (CCD). Pendrin mediates both base secretion in response to systemic base load and Cl- reabsorption in response to systemic volume depletion, manifested as decreased nephron salt and water delivery to the distal nephron. Pendrin-mediated Cl- /HCO3- exchange in the apical membrane is upregulated through stimulation of the ß-IC apical membrane G protein-coupled receptor, 2-oxoglutarate receptor 1 (OXGR1/GPR99), by its ligand α-ketoglutarate (αKG). αKG is both filtered by the glomerulus and lumenally secreted by proximal tubule apical membrane organic anion transporters (OATs). OXGR1-mediated regulation of Pendrin by αKG has been documented in transgenic mice and in isolated perfused CCD. However, aspects of the OXGR1 signaling pathway have remained little investigated since its original discovery in lymphocytes. Moreover, no ex vivo cellular system has been reported in which to study the OXGR1 signaling pathway of Type B-IC, a cell type refractory to survival in culture in its differentiated state. As Xenopus oocytes express robust heterologous Pendrin activity, we investigated OXGR1 regulation of Pendrin in oocytes. Despite functional expression of OXGR1 in oocytes, co-expression of Pendrin and OXGR1 failed to exhibit αKG-sensitive stimulation of Pendrin-mediated Cl- /anion exchange under a wide range of conditions. We conclude that Xenopus oocytes lack one or more essential molecular components or physical conditions required for OXGR1 to regulate Pendrin activity.


Assuntos
Ácidos Cetoglutáricos , Oócitos , Receptores Purinérgicos P2 , Transportadores de Sulfato , Animais , Ânions , Ácidos Cetoglutáricos/farmacologia , Camundongos , Oócitos/metabolismo , Receptores Purinérgicos P2/metabolismo , Transportadores de Sulfato/metabolismo , Xenopus laevis
5.
J Lipid Res ; 63(6): 100210, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35439525

RESUMO

Diverse molecular species of sulfatide with differences in FA lengths, unsaturation degrees, and hydroxylation statuses are expressed in the kidneys. However, the physiological functions of specific sulfatide species in the kidneys are unclear. Here, we evaluated the distribution of specific sulfatide species in the kidneys and their physiological functions. Electron microscopic analysis of kidneys of Cst-deficient mice lacking sulfatide showed vacuolar accumulation in the cytoplasm of intercalated cells in the collecting duct, whereas the proximal and distal tubules were unchanged. Immunohistochemical analysis revealed that vacuolar H+-ATPase-positive vesicles were accumulated in intercalated cells in sulfatide-deficient kidneys. Seventeen sulfatide species were detected in the murine kidney by iMScope MALDI-MS analysis. The distribution of the specific sulfatide species was classified into four patterns. Although most sulfatide species were highly expressed in the outer medullary layer, two unique sulfatide species of m/z 896.6 (predicted ceramide structure: t18:0-C22:0h) and m/z 924.6 (predicted ceramide structure: t18:0-C24:0h) were dispersed along the collecting duct, implying expression in intercalated cells. In addition, the intercalated cell-enriched fraction was purified by fluorescence-activated cell sorting using the anti-vacuolar H+-ATPase subunit 6V0A4, which predominantly contained sulfatide species (m/z 896.6 and 924.6). The Degs2 and Fa2h genes, which are responsible for ceramide hydroxylation, were expressed in the purified intercalated cells. These results suggested that sulfatide molecular species with ceramide composed of phytosphingosine (t18:0) and 2-hydroxy FAs, which were characteristically expressed in intercalated cells, were involved in the excretion of NH3 and protons into the urine.


Assuntos
Sulfoglicoesfingolipídeos , ATPases Vacuolares Próton-Translocadoras , Animais , Ceramidas , Rim/metabolismo , Camundongos , Esfingosina/análogos & derivados , ATPases Vacuolares Próton-Translocadoras/metabolismo
6.
Virchows Arch ; 478(4): 647-658, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32812119

RESUMO

FOXI1 is a forkhead family transcription factor that plays a key role in differentiation and functional maintenance for the renal intercalated cell (IC). The diagnostic utility of FOXI1 is rarely studied thus far. Comparative analyses of FOXI1 mRNA expression in normal kidney tissue and different renal neoplasms including chromophobe renal cell carcinoma (chRCC), renal oncocytoma (RO), and other renal cell carcinomas were conducted using transcriptomic data from The Cancer Genome Atlas (TCGA), Gene Expression Omnibus, and single-cell RNA-seq datasets, in combination with integrative analyses using mutant data, karyotype data, and digital slides for cases with anomalous FOXI1 expression in TCGA. Formalin-fixed, paraffin-embedded whole-tissue slides of varied primary renal neoplasms (n = 367) were subjected to FOXI1 staining for validating FOXI1 transcription levels. We confirmed that FOXI1 was significantly upregulated at mRNA levels in ICs, chRCCs, and ROs compared with other renal tubule cell and renal cell carcinoma subtypes. Furthermore, most of the cases with FOXI1 expression outliers were misclassified in the TCGA kidney cancer project. An underlying novel entity with frequent mutations involved in the mTOR pathway was also found. FOXI1 immunoreactivity was consistently noted in ICs of the distal nephron. FOXI1 staining was positive in 85 of 93 chRCCs and 13 of 18 ROs, respectively. FOXI1 staining was not seen in renal neoplasms (n = 254) derived from non-ICs. In conclusion, FOXI1 expression in normal kidney tissue is restricted to ICs. This cell type-specific expression is retained during neoplastic transformation from ICs to chRCCs or ROs. FOXI1 is thereby a potential biomarker of IC-related tumors.


Assuntos
Adenoma Oxífilo/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Renais/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Neoplasias Renais/metabolismo , Rim/metabolismo , Transcriptoma , Adenoma Oxífilo/patologia , Carcinoma de Células Renais/patologia , Estudos de Casos e Controles , Análise por Conglomerados , Humanos , Imuno-Histoquímica , Rim/patologia , Neoplasias Renais/patologia , Regulação para Cima
7.
Proc Natl Acad Sci U S A ; 117(42): 26470-26481, 2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33004624

RESUMO

The diversity and near universal expression of G protein-coupled receptors (GPCR) reflects their involvement in most physiological processes. The GPCR superfamily is the largest in the human genome, and GPCRs are common pharmaceutical targets. Therefore, uncovering the function of understudied GPCRs provides a wealth of untapped therapeutic potential. We previously identified an adhesion-class GPCR, Gpr116, as one of the most abundant GPCRs in the kidney. Here, we show that Gpr116 is highly expressed in specialized acid-secreting A-intercalated cells (A-ICs) in the kidney using both imaging and functional studies, and we demonstrate in situ receptor activation using a synthetic agonist peptide unique to Gpr116. Kidney-specific knockout (KO) of Gpr116 caused a significant reduction in urine pH (i.e., acidification) accompanied by an increase in blood pH and a decrease in pCO2 compared to WT littermates. Additionally, immunogold electron microscopy shows a greater accumulation of V-ATPase proton pumps at the apical surface of A-ICs in KO mice compared to controls. Furthermore, pretreatment of split-open collecting ducts with the synthetic agonist peptide significantly inhibits proton flux in ICs. These data suggest a tonic inhibitory role for Gpr116 in the regulation of V-ATPase trafficking and urinary acidification. Thus, the absence of Gpr116 results in a primary excretion of acid in KO mouse urine, leading to mild metabolic alkalosis ("renal tubular alkalosis"). In conclusion, we have uncovered a significant role for Gpr116 in kidney physiology, which may further inform studies in other organ systems that express this GPCR, such as the lung, testes, and small intestine.


Assuntos
Rim/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Fenômenos Bioquímicos , Transporte Biológico , Movimento Celular/fisiologia , Células Epiteliais/metabolismo , Feminino , Homeostase , Humanos , Túbulos Renais/metabolismo , Masculino , Camundongos , Camundongos Knockout
8.
Dev Biol ; 466(1-2): 1-11, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32800756

RESUMO

The distal nephron and collecting duct segments of the mammalian kidney consist of intercalated cell types intermingled among principal cell types. Notch signaling ensures that a sufficient number of cells select a principal instead of an intercalated cell fate. However, the precise mechanisms by which Notch signaling patterns the distal nephron and collecting duct cell fates is unknown. Here we observed that Hes1, a direct target of Notch signaling pathway, is required within the mouse developing collecting ducts for repression of Foxi1 expression, an essential intercalated cell specific transcription factor. Interestingly, inactivation of Foxi1 in Hes1-deficient collecting ducts rescues the deficiency in principal cell fate selection, overall urine concentrating deficiency, and reduces the occurrence of hydronephrosis. However, Foxi1 inactivation does not rescue the reduction in expression of all principal cell genes in the Hes1-deficient kidney collecting duct cells that select the principal cell fate. Additionally, suppression of Notch/Hes1 signaling in mature principal cells reduces principal cell gene expression without activating Foxi1. We conclude that Hes1 is a Notch signaling target that is essential for normal patterning of the collecting ducts with intermingled cell types by repressing Foxi1, and for maintenance of principal cell gene expression independent of repressing Foxi1.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Rim/embriologia , Receptores Notch/metabolismo , Transdução de Sinais , Fatores de Transcrição HES-1/deficiência , Animais , Fatores de Transcrição Forkhead/genética , Camundongos , Camundongos Mutantes , Receptores Notch/genética , Fatores de Transcrição HES-1/metabolismo
9.
Pflugers Arch ; 472(8): 1079-1092, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32577861

RESUMO

Fine-tuning of salt and acid-base homeostasis is achieved in the renal collecting duct through the action of intercalated and principal cells. Their activity is tightly regulated adapting to changes in systemic acid-base, fluid, or electrolyte status. The relative number of acid or bicarbonate secretory intercalated cells changes in response to acid or alkali loading. Several factors that may induce collecting duct plasticity in response to acid loading have been identified including cell proliferation, Growth Differentiation Factor 15 (Gdf15), hensin (DMBT1), and SDF1 (or CXCL12). Also, the transcription factors Foxi1 and CP2L1, or the Notch2-Jag1 signaling pathway, may play a role. However, little is known about the mechanisms mediating the adaptive response of the collecting duct to alkali loading. Here, we examined in mouse kidney the response of these factors to alkali loading. Mice were left untreated or received NaHCO3 or NaCl over 7 days. Cell proliferation in vivo was monitored by Ki67 labeling or BrdU incorporation and expression of cell markers, and regulatory factors were examined. Foxi1 and GDF15 were upregulated and CP2L1 downregulated during alkali loading. Ki67 staining and BrdU incorporation were frequent in AQP2-positive cells in the NaCl and NaHCO3 groups, but no evidence was found for increased Ki67 or BrdU staining in bicarbonate-secretory cells consistent with a model that AQP2 positive precursor cells may differentiate into intercalated cells. Thus, alkali loading alters the cellular profile of the collecting duct, which may involve cell proliferation and changes in the network of molecules determining the plasticity of the collecting duct.


Assuntos
Álcalis/metabolismo , Túbulos Renais Coletores/metabolismo , Equilíbrio Ácido-Base/fisiologia , Animais , Bicarbonatos/metabolismo , Biomarcadores/metabolismo , Proliferação de Células/fisiologia , Regulação para Baixo/fisiologia , Homeostase/fisiologia , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Regulação para Cima/fisiologia
10.
Function (Oxf) ; 1(1): zqaa007, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-35330743

RESUMO

The revolution of the omics technologies has enabled profiling of the molecules of any sample. However, the heterogeneity of the kidney with highly specialized nephron segments like the cortical collecting duct (CCD) poses a challenge regarding integration of omics data and functional analysis. We examined function and proteome from the same single CCDs of C57Bl6 mice by investigating them in a double-barreled perfusion system before targeted mass spectrometry. Transepithelial voltage (Vte), transepithelial resistance, as well as amiloride-sensitive voltage (ΔVteamil) were recorded. CCDs were of 400-600 µm of length, showed lumen negative Vte between -8.5 and -32.5 mV and an equivalent short circuit current I'sc between 54 and 192 µA/cm2. On a single-tubule proteome level, intercalated cell (IC) markers strongly correlated with other intercalated cell markers and negatively with principal cell markers. Integration of proteome data with phenotype data revealed that tubular length correlated with actin and Na+-K+-ATPase expression. ΔVte(amil) reflected the expression level of the ß-subunit of the epithelial sodium channel. Intriguingly, ΔVte(amil) correlated inversely with the water channel AQP2 and the negative regulator protein NEDD4L (NEDD4-2). In pendrin knockout (KO) mice, the CCD proteome was accompanied by strong downregulation of other IC markers like CLCNKB, BSND (Barttin), and VAA (vH+-ATPase), a configuration that may contribute to the salt-losing phenotype of Pendred syndrome. Proteins normally coexpressed with pendrin were decreased in pendrin KO CCDs. In conclusion, we show that functional proteomics on a single nephron segment scale allows function-proteome correlations, and may potentially help predicting function from omics data.


Assuntos
Túbulos Renais Coletores , Animais , Camundongos , Aquaporina 2/genética , Proteoma/genética , Proteômica , Camundongos Endogâmicos C57BL , Transportadores de Sulfato/genética , Fenótipo , Adenosina Trifosfatases/genética
11.
Am J Physiol Renal Physiol ; 315(4): F812-F823, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28468965

RESUMO

The urinary tract is usually culture negative despite its close proximity to microbial flora. The precise mechanism by which the kidneys and urinary tract defends against infection is not well understood. The initial kidney cells to encounter ascending pathogens are the collecting tubule cells that consist of principal cells (PCs) that express aquaporin 2 (AQP2) and intercalated cells (ICs) that express vacuolar H+-ATPase (V-ATPase, B1 subunit). We have previously shown that ICs are involved with the human renal innate immune defense. Here we generated two reporter mice, VATPase B1-cre+tdT+ mice to fluorescently label ICs and AQP2-cre+tdT+ mice to fluorescently label PCs, and then performed flow sorting to enrich PCs and ICs for analysis. Isolated ICs and PCs along with proximal tubular cells were used to measure antimicrobial peptide (AMP) mRNA expression. ICs and PCs were significantly enriched for AMPs. Isolated ICs responded to uropathogenic Escherichia coli (UPEC) challenge in vitro and had higher RNase4 gene expression than control while both ICs and PCs responded to UPEC challenge in vivo by upregulating Defb1 mRNA expression. To our knowledge, this is the first report of isolating murine collecting tubule cells and performing targeted analysis for multiple classes of AMPs.


Assuntos
Aquaporina 2/imunologia , Células Epiteliais/metabolismo , Túbulos Renais Coletores/imunologia , Reação em Cadeia da Polimerase , Animais , Aquaporina 2/genética , Imunidade Inata/imunologia , Rim/imunologia , Rim/metabolismo , Camundongos Transgênicos , Reação em Cadeia da Polimerase/métodos , Regulação para Cima/imunologia , ATPases Vacuolares Próton-Translocadoras/imunologia , ATPases Vacuolares Próton-Translocadoras/metabolismo
12.
Proc Natl Acad Sci U S A ; 114(46): E9989-E9998, 2017 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-29089413

RESUMO

Prior RNA sequencing (RNA-seq) studies have identified complete transcriptomes for most renal epithelial cell types. The exceptions are the cell types that make up the renal collecting duct, namely intercalated cells (ICs) and principal cells (PCs), which account for only a small fraction of the kidney mass, but play critical physiological roles in the regulation of blood pressure, extracellular fluid volume, and extracellular fluid composition. To enrich these cell types, we used FACS that employed well-established lectin cell surface markers for PCs and type B ICs, as well as a newly identified cell surface marker for type A ICs, c-Kit. Single-cell RNA-seq using the IC- and PC-enriched populations as input enabled identification of complete transcriptomes of A-ICs, B-ICs, and PCs. The data were used to create a freely accessible online gene-expression database for collecting duct cells. This database allowed identification of genes that are selectively expressed in each cell type, including cell-surface receptors, transcription factors, transporters, and secreted proteins. The analysis also identified a small fraction of hybrid cells expressing aquaporin-2 and anion exchanger 1 or pendrin transcripts. In many cases, mRNAs for receptors and their ligands were identified in different cells (e.g., Notch2 chiefly in PCs vs. Jag1 chiefly in ICs), suggesting signaling cross-talk among the three cell types. The identified patterns of gene expression among the three types of collecting duct cells provide a foundation for understanding physiological regulation and pathophysiology in the renal collecting duct.


Assuntos
Aquaporina 2/metabolismo , Células Epiteliais/metabolismo , Túbulos Renais Coletores/metabolismo , Rim/metabolismo , Análise de Sequência de RNA/métodos , Transcriptoma , Animais , Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Proteínas de Transporte de Ânions/metabolismo , Sequência de Bases , Biomarcadores/metabolismo , Expressão Gênica , Perfilação da Expressão Gênica , Proteína Jagged-1/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , RNA/metabolismo , Receptor Notch2/metabolismo , Transdução de Sinais , Transportadores de Sulfato , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma/genética
13.
Dev Biol ; 424(1): 77-89, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28215940

RESUMO

The mammalian kidney collecting ducts are critical for water, electrolyte and acid-base homeostasis and develop as a branched network of tubular structures composed of principal cells intermingled with intercalated cells. The intermingled nature of the different collecting duct cell types has made it challenging to identify unique and critical factors that mark and/or regulate the development of the different collecting duct cell lineages. Here we report that the canonical Notch signaling pathway components, RBPJ and Presinilin1 and 2, are involved in patterning the mouse collecting duct cell fates by maintaining a balance between principal cell and intercalated cell fates. The relatively reduced number of principal cells in Notch-signaling-deficient kidneys offered a unique genetic leverage to identify critical principal cell-enriched factors by transcriptional profiling. Elf5, which codes for an ETS transcription factor, is one such gene that is down-regulated in kidneys with Notch-signaling-deficient collecting ducts. Additionally, Elf5 is among the earliest genes up regulated by ectopic expression of activated Notch1 in the developing collecting ducts. In the kidney, Elf5 is first expressed early within developing collecting ducts and remains on in mature principal cells. Lineage tracing of Elf5-expressing cells revealed that they are committed to the principal cell lineage by as early as E16.5. Over-expression of ETS Class IIa transcription factors, including Elf5, Elf3 and Ehf, increase the transcriptional activity of the proximal promoters of Aqp2 and Avpr2 in cultured ureteric duct cell lines. Conditional inactivation of Elf5 in the developing collecting ducts results in a small but significant reduction in the expression levels of Aqp2 and Avpr2 genes. We have identified Elf5 as an early maker of the principal cell lineage that contributes to the expression of principal cell specific genes.


Assuntos
Aquaporina 2/genética , Linhagem da Célula , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Rim/citologia , Rim/metabolismo , Receptores de Vasopressinas/genética , Fatores de Transcrição/metabolismo , Animais , Aquaporina 2/metabolismo , Contagem de Células , Linhagem Celular , Regulação para Baixo/genética , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Integrases/metabolismo , Rim/embriologia , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/embriologia , Túbulos Renais Coletores/metabolismo , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , Receptores Notch/metabolismo , Receptores de Vasopressinas/metabolismo , Transdução de Sinais , Regulação para Cima/genética , Ureter/embriologia , Ureter/metabolismo
14.
Am J Physiol Renal Physiol ; 312(1): F143-F156, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27806944

RESUMO

Within the CCD of the distal nephron of the rabbit, the BK (maxi K) channel mediates Ca2+- and/or stretch-dependent flow-induced K+ secretion (FIKS) and contributes to K+ adaptation in response to dietary K+ loading. An unresolved question is whether BK channels in intercalated cells (ICs) and/or principal cells (PCs) in the CCD mediate these K+ secretory processes. In support of a role for ICs in FIKS is the higher density of immunoreactive apical BKα (pore-forming subunit) and functional BK channel activity than detected in PCs, and an increase in IC BKα expression in response to a high-K+ diet. PCs possess a single apical cilium which has been proposed to serve as a mechanosensor; direct manipulation of cilia leads to increases in cell Ca2+ concentration, albeit of nonciliary origin. Immunoperfusion of isolated and fixed CCDs isolated from control K+-fed rabbits with channel subunit-specific antibodies revealed colocalization of immunodetectable BKα- and ß1-subunits in cilia as well as on the apical membrane of cilia-expressing PCs. Ciliary BK channels were more easily detected in rabbits fed a low-K+ vs. high-K+ diet. Single-channel recordings of cilia revealed K+ channels with conductance and kinetics typical of the BK channel. The observations that 1) FIKS was preserved but 2) the high-amplitude Ca2+ peak elicited by flow was reduced in microperfused CCDs subject to pharmacological deciliation suggest that cilia BK channels do not contribute to K+ secretion in this segment, but that cilia serve as modulators of cell signaling.


Assuntos
Cálcio/metabolismo , Cílios/metabolismo , Túbulos Renais Coletores/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Néfrons/metabolismo , Potássio/metabolismo , Animais , Feminino , Córtex Renal/metabolismo , Coelhos
15.
Clin Exp Nephrol ; 20(5): 663-670, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26846783

RESUMO

The aldosterone-mineralocorticoid receptor (MR) system serves as the major regulator of fluid homeostasis, and is an important drug target for the treatment of hypertension, heart failure, and chronic kidney disease. While the ligand aldosterone plays a central role in facilitating MR activity, recent studies have revealed that MR signaling is modulated through distinct mechanisms at the levels of the receptor and the downstream targets. Notably, phosphorylation of the ligand-binding domain in MR regulates the ability of the receptor to bind to ligand in renal intercalated cells, providing an additional layer of regulation that allows the cell-selective control of MR signaling. These mechanisms are involved in the context-dependent effects of aldosterone in the distal nephron. In this article, the recent progress in the understanding of mechanisms regulating the action of aldosterone is discussed, focusing on the connecting tubules and collecting duct in the kidney.


Assuntos
Aldosterona/metabolismo , Rim/metabolismo , Receptores de Mineralocorticoides/metabolismo , Sistema Renina-Angiotensina , Transdução de Sinais , Animais , Humanos , Ligantes , Fosforilação , Potássio/metabolismo , Ligação Proteica , Cloreto de Sódio/metabolismo
16.
Front Behav Neurosci ; 9: 249, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26441578

RESUMO

Altered medial prefrontal cortex (mPFC) and amygdala function is associated with anxiety-related disorders. While the mPFC-amygdala pathway has a clear role in fear conditioning, these structures are also involved in active avoidance. Given that avoidance perseveration represents a core symptom of anxiety disorders, the neural substrate of avoidance, especially its extinction, requires better understanding. The present study was designed to investigate the activity, particularly, inhibitory neuronal activity in mPFC and amygdala during acquisition and extinction of lever-press avoidance in rats. Neural activity was examined in the mPFC, intercalated cell clusters (ITCs) lateral (LA), basal (BA) and central (CeA) amygdala, at various time points during acquisition and extinction, using induction of the immediate early gene product, c-Fos. Neural activity was greater in the mPFC, LA, BA, and ITC during the extinction phase as compared to the acquisition phase. In contrast, the CeA was the only region that was more activated during acquisition than during extinction. Our results indicate inhibitory neurons are more activated during late phase of acquisition and extinction in the mPFC and LA, suggesting the dynamic involvement of inhibitory circuits in the development and extinction of avoidance response. Together, these data start to identify the key brain regions important in active avoidance behavior, areas that could be associated with avoidance perseveration in anxiety disorders.

17.
Am J Physiol Renal Physiol ; 309(5): F464-73, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26084929

RESUMO

During metabolic acidosis, the cortical collecting duct (CCD) of the rabbit reverses the polarity of bicarbonate flux from net secretion to net absorption, and this is accomplished by increasing the proton secretory rate by α-intercalated cells (ICs) and decreasing bicarbonate secretion by ß-ICs. To better characterize dynamic changes in H(+)-secreting α-ICs, we examined their morphology in collecting ducts microdissected from kidneys of normal, acidotic, and recovering rabbits. α-ICs in defined axial regions varied in number and basolateral anion exchanger (AE)1 morphology, which likely reflects their relative activity and function along the collecting duct. Upon transition from CCD to outer medullary collecting duct from the outer stripe to the inner stripe, the number of α-ICs increases from 11.0 ± 1.2 to 15.4 ± 1.11 and to 32.0 ± 1.3 cells/200 µm, respectively. In the CCD, the basolateral structure defined by AE1 typically exhibited a pyramidal or conical shape, whereas in the medulla the morphology was elongated and shallow, resulting in a more rectangular shape. Furthermore, acidosis reversibly induced α-ICs in the CCD to acquire a more rectangular morphology concomitant with a transition from diffusely cytoplasmic to increased basolateral surface distribution of AE1 and apical polarization of B1-V-ATPase. The latter results are consistent with the supposition that morphological adaptation from the pyramidal to rectangular shape reflects a transition toward a more "active" configuration. In addition, α-ICs in the outer medullary collecting duct from the outer stripe exhibited cellular morphology strikingly similar to dendritic cells that may reflect a newly defined ancillary function in immune defense of the kidney.


Assuntos
Acidose/patologia , Forma Celular/fisiologia , Túbulos Renais Coletores/patologia , Acidose/metabolismo , Animais , Feminino , Túbulos Renais Coletores/metabolismo , Coelhos
18.
Clin Exp Nephrol ; 19(5): 771-82, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25500736

RESUMO

BACKGROUND: The localization and role of the calcium-sensing receptor (CaSR) along the nephron including the collecting ducts is still open to debate. METHODS: Using the quantitative, highly sensitive in situ hybridization technique and a double-staining immunohistochemistry technique, we investigated the axial distribution and expression of CaSR along the nephron in mice (C57B/6J) treated for 6 days with acid or alkali diets. RESULTS: Under control condition, CaSR was specifically localized in the cortical and medullary thick ascending limb of Henle's loop (CTAL and MTAL), macula densa (MD), distal convoluted tubule (DCT), and CCD (TALs, MD > DCT, CCD). Along the CCD, CaSR was co-localized with an anion exchanger type 4 (AE4), a marker of the basolateral membrane of type-B intercalated cell (IC-B) in mice. On the contrary, CaSR was not detected either in principal cells (PC) or in type-A intercalated cell (IC-A). CaSR expression levels in IC-B significantly (P < 0.005) decreased when mice were fed NH4Cl (acid) diets and increased when animals were given NaHCO3 (alkali) diets. As expected, cell heights of IC-A and IC-B significantly (P < 0.005) increased in the above experimental conditions. Surprisingly, single infusion (ip) of neomycin, an agonist of CaSR, significantly (P < 0.005) increased urinary Ca excretion without further increasing the hourly urine volume and significantly (P < 0.05) decreased urine pH. CONCLUSION: CaSR, cloned from rat kidney, was localized in the basolateral membrane of IC-B and was more expressed during alkali-loading. Its alkali-sensitive expression may promote urinary alkali secretion for body acid-base balance.


Assuntos
Túbulos Renais Coletores/metabolismo , Rim/metabolismo , Receptores Acoplados a Proteínas G/biossíntese , Animais , Cálcio/urina , Tamanho Celular , Diuréticos/farmacologia , Concentração de Íons de Hidrogênio , Hibridização In Situ , Rim/citologia , Túbulos Renais Coletores/citologia , Camundongos , Camundongos Endogâmicos C57BL , Néfrons/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Detecção de Cálcio , Receptores Acoplados a Proteínas G/genética
19.
Am J Physiol Renal Physiol ; 306(10): F1107-20, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24647713

RESUMO

Renal ammonia metabolism is a fundamental element of acid-base homeostasis, comprising a major component of both basal and physiologically altered renal net acid excretion. Over the past several years, a fundamental change in our understanding of the mechanisms of renal epithelial cell ammonia transport has occurred, replacing the previous model which was based upon diffusion equilibrium for NH3 and trapping of NH4(+) with a new model in which specific and regulated transport of both NH3 and NH4(+) across renal epithelial cell membranes via specific membrane proteins is required for normal ammonia metabolism. A major advance has been the recognition that members of a recently recognized transporter family, the Rhesus glycoprotein family, mediate critical roles in renal and extrarenal ammonia transport. The erythroid-specific Rhesus glycoprotein, Rh A Glycoprotein (Rhag), was the first Rhesus glycoprotein recognized as an ammonia-specific transporter. Subsequently, the nonerythroid Rh glycoproteins, Rh B Glycoprotein (Rhbg) and Rh C Glycoprotein (Rhcg), were cloned and identified as ammonia transporters. They are expressed in specific cell populations and membrane domains in distal renal epithelial cells, where they facilitate ammonia secretion. In this review, we discuss the distribution of Rhbg and Rhcg in the kidney, the regulation of their expression and activity in physiological disturbances, the effects of genetic deletion on renal ammonia metabolism, and the molecular mechanisms of Rh glycoprotein-mediated ammonia transport.


Assuntos
Amônia/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Glicoproteínas/metabolismo , Rim/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Equilíbrio Ácido-Base/fisiologia , Animais , Transporte Biológico/fisiologia , Homeostase/fisiologia , Humanos , Camundongos , Modelos Animais
20.
Am J Physiol Renal Physiol ; 305(6): F919-29, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23825070

RESUMO

The cellular morphology of the collecting duct is altered by chronic lithium treatment. We have previously shown that lithium increases the fraction of type-A intercalated cells and lowers the fraction of principal cells along the collecting duct. Moreover, type-A intercalated cells acquire a long-row distribution pattern along the tubules. In the present study, we show that these morphological changes reverse progressively after discontinuation of lithium and finally disappear after 19 days from lithium suspension. In this time frame we have identified for the first time, in vivo, a novel cellular type positive for both intercalated and principal cells functional markers, as recognized by colabeling with H(+)-ATPase/aquaporin-4 (AQP4) and anion exchanger-1 (AE-1)/AQP2 and Foxi1/AQP4. This cell type is mainly present after 6 days of lithium washout, and it disappears in parallel with the long-row pattern of the type-A intercalated cells. It usually localizes either in the middle or at the edge of the long-row pattern. Its ultrastructure resembles the intercalated cells as shown both by differential interference contrast and by electron microscopy. The time course of appearance, the localization along the collecting duct, and the ultrastructure suggest that the cells double labeled for principal and intercalated cells markers could represent a transition element driving the conversion of intercalated cells into principal cells.


Assuntos
Diabetes Insípido Nefrogênico/induzido quimicamente , Diabetes Insípido Nefrogênico/patologia , Túbulos Renais Coletores/citologia , Lítio/efeitos adversos , Síndrome de Abstinência a Substâncias , Animais , Aquaporina 2/biossíntese , Aquaporina 4/biossíntese , Proliferação de Células , Diabetes Insípido Nefrogênico/metabolismo , Fatores de Transcrição Forkhead/biossíntese , Masculino , ATPases Translocadoras de Prótons/biossíntese , Ratos , Síndrome de Abstinência a Substâncias/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA