Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
PLoS One ; 15(12): e0244251, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33332450

RESUMO

The connexin 37 (Cx37) channel is clustered at gap junctions between cells in the renal vasculature or the renal tubule where it is abundant in basolateral cell interdigitations and infoldings of epithelial cells in the proximal tubule, thick ascending limb, distal convoluted tubule and collecting duct; however, physiological data regarding its role are limited. In this study, we investigated the role of Cx37 in fluid homeostasis using mice with a global deletion of Cx37 (Cx37-/- mice). Under baseline conditions, Cx37-/- had ~40% higher fluid intake associated with ~40% lower urine osmolality compared to wild-type (WT) mice. No differences were observed between genotypes in urinary adenosine triphosphate or prostaglandin E2, paracrine factors that alter renal water handling. After 18-hours of water deprivation, plasma aldosterone and urine osmolality increased significantly in Cx37-/- and WT mice; however, the latter remained ~375 mmol/kg lower in Cx37-/- mice, an effect associated with a more pronounced body weight loss despite higher urinary AVP/creatinine ratios compared to WT mice. Consistent with this, fluid intake in the first 3 hours after water deprivation was 37% greater in Cx37-/- vs WT mice. Cx37-/- mice showed significantly lower renal AQP2 abundance and AQP2 phosphorylation at serine 256 than WT mice in response to vehicle or dDAVP, suggesting a partial contribution of the kidney to the lower urine osmolality. The abundance and responses of the vasopressin V2 receptor, AQP3, NHE3, NKCC2, NCC, H+-ATPase, αENaC, γENaC or Na+/K+-ATPase were not significantly different between genotypes. In summary, these results demonstrate that Cx37 is important for body water handling.


Assuntos
Aquaporina 2/metabolismo , Conexinas/fisiologia , Deleção de Genes , Polidipsia/etiologia , Poliúria/etiologia , Animais , Aquaporina 2/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Polidipsia/patologia , Poliúria/patologia , Proteína alfa-4 de Junções Comunicantes
2.
Cells ; 9(10)2020 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-32993088

RESUMO

As a rare hereditary disease, congenital nephrogenic diabetes insipidus (NDI) is clinically characterized by polyuria with hyposthenuria and polydipsia. NDI results from collecting duct principal cell hyporesponsiveness or insensitivity to the antidiuretic action of arginine vasopressin (AVP). The principal cell-specific water channel aquaporin-2 (AQP2) plays an essential role in water reabsorption along osmotic gradients. The capacity to accumulate AQP2 in the apical plasma membrane in response to decreased fluid volume or increased plasma osmolality is critically regulated by the antidiuretic hormone AVP and its receptor 2 (AVPR2). Mutations in AVPR2 result in X-linked recessive NDI, the most common form of inherited NDI. Genetic defects in AQP2 cause autosomal recessive or dominant NDI. In this review, we provide an updated overview of the genetic and molecular mechanisms of congenital NDI, with a focus on the potential disease-causing mutations in AVPR2 and AQP2, the molecular defects in the AVPR2 and AQP2 mutants, post-translational modifications (i.e., phosphorylation, ubiquitination, and glycosylation) and various protein-protein interactions that regulate phosphorylation, ubiquitination, tetramerization, trafficking, stability, and degradation of AQP2.


Assuntos
Aquaporina 2/genética , Diabetes Insípido Nefrogênico/genética , Neurofisinas/genética , Precursores de Proteínas/genética , Receptores de Vasopressinas/genética , Vasopressinas/genética , Diabetes Insípido Nefrogênico/patologia , Humanos , Mutação/genética , Poliúria/genética , Poliúria/patologia , Mapas de Interação de Proteínas/genética , Processamento de Proteína Pós-Traducional/genética , Transporte Proteico/genética
3.
Am J Physiol Renal Physiol ; 318(5): F1199-F1209, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32249612

RESUMO

Lithium is widely used in psychiatry as the golden standard for more than 60 yr due to its effectiveness. However, its adverse effect has been limiting its long-term use in clinic. About 40% of patients taking lithium develop nephrogenic diabetes insipidus (NDI). Lithium can also induce proliferation of collecting duct cells, leading to microcyst formation in the kidney. Lithium was considered an autophagy inducer that might contribute to the therapeutic benefit of neuropsychiatric disorders. Thus, we hypothesized that autophagy may play a role in lithium-induced kidney nephrotoxicity. To address our hypothesis, we fed mice with a lithium-containing diet with chloroquine (CQ), an autophagy inhibitor, concurrently. Lithium-treated mice presented enhanced autophagy activity in the kidney cortex and medulla. CQ treatment significantly ameliorated lithium-induced polyuria, polydipsia, natriuresis, and kaliuresis accompanied with attenuated downregulation of aquaporin-2 and Na+-K+-2Cl- cotransporter protein. The protective effect of CQ on aquaporin-2 protein abundance was confirmed in cultured cortical collecting duct cells. In addition, we found that lithium-induced proliferation of collecting duct cells was also suppressed by CQ as detected by proliferating cell nuclear antigen staining. Moreover, both phosphorylated mammalian target of rapamycin and ß-catenin expression, which have been reported to be increased by lithium and associated with cell proliferation, were reduced by CQ. Taken together, our study demonstrated that CQ protected against lithium-induced NDI and collecting duct cell proliferation possibly through inhibiting autophagy.


Assuntos
Proliferação de Células/efeitos dos fármacos , Cloroquina/farmacologia , Diabetes Insípido Nefrogênico/prevenção & controle , Túbulos Renais Coletores/efeitos dos fármacos , Cloreto de Lítio , Animais , Aquaporina 2/genética , Aquaporina 2/metabolismo , Autofagia/efeitos dos fármacos , Linhagem Celular , Diabetes Insípido Nefrogênico/induzido quimicamente , Diabetes Insípido Nefrogênico/metabolismo , Diabetes Insípido Nefrogênico/patologia , Dinoprostona/urina , Modelos Animais de Doenças , Túbulos Renais Coletores/metabolismo , Túbulos Renais Coletores/patologia , Masculino , Camundongos da Linhagem 129 , Natriurese/efeitos dos fármacos , Fosforilação , Poliúria/induzido quimicamente , Poliúria/metabolismo , Poliúria/patologia , Poliúria/prevenção & controle , Membro 1 da Família 12 de Carreador de Soluto/genética , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , beta Catenina/metabolismo
4.
Am J Physiol Renal Physiol ; 317(6): F1450-F1461, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31566426

RESUMO

Diabetic kidney disease (DKD) is one of the leading pathological causes of decreased renal function and progression to end-stage kidney failure. To explore and characterize age-related changes in DKD and associated glomerular damage, we used a rat model of type 2 diabetic nephropathy (T2DN) at 12 wk and older than 48 wk. We compared their disease progression with control nondiabetic Wistar and diabetic Goto-Kakizaki (GK) rats. During the early stages of DKD, T2DN and GK animals revealed significant increases in blood glucose and kidney-to-body weight ratio. Both diabetic groups had significantly altered renin-angiotensin-aldosterone system function. Thereafter, during the later stages of disease progression, T2DN rats demonstrated a remarkable increase in renal damage compared with GK and Wistar rats, as indicated by renal hypertrophy, polyuria accompanied by a decrease in urine osmolarity, high cholesterol, a significant prevalence of medullary protein casts, and severe forms of glomerular injury. Urinary nephrin shedding indicated loss of the glomerular slit diaphragm, which also correlates with the dramatic elevation in albuminuria and loss of podocin staining in aged T2DN rats. Furthermore, we used scanning ion microscopy topographical analyses to detect and quantify the pathological remodeling in podocyte foot projections of isolated glomeruli from T2DN animals. In summary, T2DN rats developed renal and physiological abnormalities similar to clinical observations in human patients with DKD, including progressive glomerular damage and a significant decrease in renin-angiotensin-aldosterone system plasma levels, indicating these rats are an excellent model for studying the progression of renal damage in type 2 DKD.


Assuntos
Diabetes Mellitus Tipo 2/patologia , Nefropatias Diabéticas/patologia , Envelhecimento , Albuminúria/etiologia , Albuminúria/prevenção & controle , Animais , Glicemia/metabolismo , Progressão da Doença , Hipertrofia , Glomérulos Renais/patologia , Masculino , Proteínas de Membrana/urina , Tamanho do Órgão , Poliúria/etiologia , Poliúria/patologia , Ratos , Ratos Wistar , Sistema Renina-Angiotensina , Desequilíbrio Hidroeletrolítico/etiologia , Desequilíbrio Hidroeletrolítico/metabolismo
5.
J Diabetes Res ; 2019: 8153140, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31467929

RESUMO

A variety of animal models of diabetes mellitus (DM) are required to study the genetics and pathophysiology of DM. We established a novel rat strain showing nonobese type 2 diabetes with enlarged kidneys from the LEA.PET-pet congenic strain and named it Diabetes with Enlarged Kidney (DEK). The body growth of DEK affected rats was similar to that of normal rats before the development of DM but was attenuated with the deterioration of DM. There was a marked difference in the etiology of DEK by gender: DM phenotypes including polyuria, polydipsia, and hyperglycemia (nonfasting blood glucose over 300 mg/dl) were found in male rats aged over 10 weeks but not in female rats. The cumulative incidence of DM in DEK males at the age of 30 weeks was 44.8%. Oral glucose tolerance tests showed glucose intolerance and decreased insulin secretion in response to glucose loading in affected males, features which were exacerbated with age. Affected males exhibited disorganized architecture of pancreatic islets, decreased numbers of ß cells, and markedly decreased expression of insulin, despite no pathological findings of hemorrhage or infiltration of inflammatory cells in the pancreatic islet. Age-related islet fibrosis appeared similar in normal and affected males. Affected males also showed enlarged kidneys with dilation of renal tubules in both the cortex and medulla, but no obvious glomerular lesions typical of diabetic nephropathy (DN) at the age of 30 weeks. Plasma levels of urea nitrogen and creatinine were normal, but hypoalbuminemia was detected. These pathophysiological features in affected males indicated that their renal function was almost maintained despite severe DM. Taken together, these findings indicate that the affected males of the DEK strain are a novel nonobese type 2 diabetes rat model useful for studying the mechanisms underlying ß cell loss and identifying genetic factors protective against DN.


Assuntos
Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/patologia , Nefropatias Diabéticas/etiologia , Rim/patologia , Animais , Animais Congênicos , Glicemia/análise , Glicemia/metabolismo , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Nefropatias Diabéticas/sangue , Nefropatias Diabéticas/patologia , Feminino , Teste de Tolerância a Glucose , Hiperglicemia/complicações , Hiperglicemia/patologia , Hipertrofia/sangue , Hipertrofia/etiologia , Masculino , Polidipsia/etiologia , Polidipsia/patologia , Poliúria/etiologia , Poliúria/patologia , Ratos , Ratos Endogâmicos
6.
Am J Physiol Renal Physiol ; 315(2): F313-F322, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29667915

RESUMO

Prolonged lithium treatment is associated with various renal side effects and is known to induce inner medullary collecting duct (IMCD) remodeling. In animals treated with lithium, the fraction of intercalated cells (ICs), which are responsible for acid-base homeostasis, increases compared with renal principal cells (PCs). To investigate the intricacies of lithium-induced IMCD remodeling, male Sprague-Dawley rats were fed a lithium-enriched diet for 0,1, 2, 3, 6, 9, or 12 wk. Urine osmolality was decreased at 1 wk, and from 2 to 12 wk, animals were severely polyuric. After 6 wk of lithium treatment, approximately one-quarter of the cells in the initial IMCD expressed vacuolar H+-ATPase, an IC marker. These cells were localized in portions of the inner medulla, where ICs are not normally found. Pendrin, a Cl-/[Formula: see text] exchanger, is normally expressed only in two IC subtypes found in the convoluted tubule, the cortical collecting duct, and the connecting tubule. At 6 wk of lithium treatment, we observed various patterns of pendrin localization and expression in the rat IMCD, including a novel phenotype wherein pendrin was coexpressed with aquaporin-4. These observations collectively suggest that renal IMCD cell plasticity may play an important role in lithium-induced IMCD remodeling.


Assuntos
Plasticidade Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Antiportadores de Cloreto-Bicarbonato/metabolismo , Túbulos Renais Coletores/efeitos dos fármacos , Carbonato de Lítio/toxicidade , Transportadores de Sulfato/metabolismo , Compostos de Amônio/urina , Animais , Aquaporina 4/metabolismo , Antiportadores de Cloreto-Bicarbonato/genética , Esquema de Medicação , Regulação da Expressão Gênica , Concentração de Íons de Hidrogênio , Túbulos Renais Coletores/metabolismo , Túbulos Renais Coletores/patologia , Masculino , Concentração Osmolar , Fenótipo , Poliúria/induzido quimicamente , Poliúria/patologia , Poliúria/urina , Ratos Sprague-Dawley , Transdução de Sinais , Transportadores de Sulfato/genética , Fatores de Tempo , ATPases Vacuolares Próton-Translocadoras/metabolismo
7.
FASEB J ; 32(8): 4544-4559, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29553830

RESUMO

A main feature of Fabry disease is nephropathy, with polyuria an early manifestation; however, the mechanism that underlies polyuria and affected tubules is unknown. To increase globotriaosylceramide (Gb3) levels, we previously crossbred asymptomatic Glatm mice with transgenic mice that expressed human Gb3 synthase (A4GALT) and generated the GlatmTg(CAG-A4GALT) symptomatic Fabry model mice. Additional analyses revealed that these mice exhibit polyuria and renal dysfunction without remarkable glomerular damage. In the present study, we investigated the mechanism of polyuria and renal dysfunction in these mice. Gb3 accumulation was mostly detected in the medulla; medullary thick ascending limbs (mTALs) were the most vacuolated tubules. mTAL cells contained lamellar bodies and had lost their characteristic structure ( i.e., extensive infolding and numerous elongated mitochondria). Decreased expression of the major molecules-Na+-K+-ATPase, uromodulin, and Na+-K+-2Cl- cotransporter-that are involved in Na+ reabsorption in mTALs and the associated loss of urine-concentrating ability resulted in progressive water- and salt-loss phenotypes. GlatmTg(CAG-A4GALT) mice exhibited fibrosis around mTALs and renal dysfunction. These and other features were consistent with pathologic findings in patients with Fabry disease. Results demonstrate that mTAL dysfunction causes polyuria and renal impairment and contributes to the pathophysiology of Fabry nephropathy.-Maruyama, H., Taguchi, A., Nishikawa, Y., Guili, C., Mikame, M., Nameta, M., Yamaguchi, Y., Ueno, M., Imai, N., Ito, Y., Nakagawa, T., Narita, I., Ishii, S. Medullary thick ascending limb impairment in the GlatmTg(CAG-A4GALT) Fabry model mice.


Assuntos
Doença de Fabry/patologia , Nefropatias/patologia , Medula Renal/patologia , Animais , Modelos Animais de Doenças , Doença de Fabry/metabolismo , Capacidade de Concentração Renal/fisiologia , Nefropatias/metabolismo , Medula Renal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Poliúria/metabolismo , Poliúria/patologia , Sódio/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Triexosilceramidas/metabolismo
8.
Am J Physiol Renal Physiol ; 315(2): F199-F210, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29592526

RESUMO

The molecular mechanisms of melamine-induced renal toxicity have not been fully understood. The purpose of the study aimed to investigate whether melamine and cyanuric acid induced NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation in the kidney, which may contribute to abnormal water and sodium handling in a rat model. Wistar rats received melamine (Mel; 200 mg·kg body wt-1·day-1), cyanuric acid (CA; 200 mg·kg body wt-1·day-1), or Mel plus CA (Mel + CA; 100 mg·kg body wt-1·day-1, each) for 2 wk. Mel + CA caused damaged tubular epithelial structure and organelles, dilated tubular lumen, and inflammatory responses. Crystals were observed in urine and serum specimen, also in the lumen of dilated distal renal tubules. The combined ingestion of Mel and CA in rats caused a markedly impaired urinary concentration, which was associated with reduced protein expression of aquaporin (AQP)1, 2, and 3 in inner medulla and α-Na-K-ATPase and Na-K-2Cl transporters in cortex and outer medulla. Mel + CA treatment was associated with increased protein expression of CD3 and mRNA levels of CD68 and F4/80 as well as phosphorylation of NF-κB in the kidney. Mel + CA treatment increased protein and mRNA expression of NLRP3 inflammasome components apoptosis-associated speck-like protein containing a caspase recruitment domain, caspase-1, and IL-1ß in the inner medulla of rats. NF-κB inhibitor Bay 11-7082 reduced IL-1ß expression induced by Mel + CA and prevented downregulation of AQP2 in inner medullary collecting duct cell suspensions. In conclusion, Mel + CA treatment caused urinary-concentrating defects and reduced expression of renal AQPs and key sodium transporters, which is likely due to the inflammatory responses and activation of NLRP3 inflammasome induced by crystals formed in the kidney.


Assuntos
Inflamassomos/metabolismo , Rim/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Poliúria/metabolismo , Triazinas , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Antígenos de Diferenciação Mielomonocítica/genética , Antígenos de Diferenciação Mielomonocítica/metabolismo , Aquaporinas/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Complexo CD3/metabolismo , Caspase 1/metabolismo , Interleucina-1beta/metabolismo , Rim/patologia , Rim/fisiopatologia , Capacidade de Concentração Renal , Masculino , NF-kappa B/metabolismo , Fosforilação , Poliúria/induzido quimicamente , Poliúria/patologia , Poliúria/fisiopatologia , Ratos Wistar , Transdução de Sinais , Simportadores de Cloreto de Sódio-Potássio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo
9.
FASEB J ; 32(2): 693-702, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28970255

RESUMO

Methionine restriction (MR) extends the lifespan across several species, such as rodents, fruit flies, roundworms, and yeast. MR studies have been conducted on various rodent organs, such as liver, adipose tissue, heart, bones, and skeletal muscle, to elucidate its benefits to the healthspan; however, studies of the direct effect of MR on kidneys are lacking. To investigate the renal effects of MR, we used young and aged unilateral nephrectomized and 5/6 nephrectomized (5/6Nx) mice. Our studies indicated that MR mice experienced polydipsia and polyuria compared with control-fed counterparts. Urine albumin, creatinine, albumin-to-creatinine ratio, sulfur amino acids, and electrolytes were reduced in MR mice. Kidneys of MR mice up-regulated genes that are involved in ion transport, such as Aqp2, Scnn1a, and Slc6a19, which indicated a response to maintain osmotic balance. In addition, we identified renoprotective biomarkers that are affected by MR, such as clusterin and cystatin C. Of importance, MR attenuated kidney injury in 5/6Nx mice by down-regulating inflammation and fibrosis mechanisms. Thus, our studies in mice show the important role of kidneys during MR in maintaining osmotic homeostasis. Moreover, our studies also show that the MR diet delays the progression of kidney disease.-Cooke, D., Ouattara, A., Ables, G. P. Dietary methionine restriction modulates renal response and attenuates kidney injury in mice.


Assuntos
Injúria Renal Aguda/metabolismo , Rim/metabolismo , Metionina/deficiência , Polidipsia/metabolismo , Poliúria/metabolismo , Injúria Renal Aguda/dietoterapia , Injúria Renal Aguda/patologia , Animais , Rim/patologia , Masculino , Camundongos , Osmose , Polidipsia/dietoterapia , Polidipsia/patologia , Poliúria/dietoterapia , Poliúria/patologia
10.
Am J Physiol Renal Physiol ; 313(4): F1026-F1037, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28701310

RESUMO

The renal collecting duct (CD) contains two major cell types, intercalated (ICs) and principal cells (PCs). A previous report showed that deletion of ß1-integrin in the entire renal CD causes defective CD morphogenesis resulting in kidney dysfunction. However, subsequent deletion of ß1-integrin specifically in ICs and PCs, respectively, did not cause any morphological defects in the CDs. The discrepancy between these studies prompts us to reinvestigate the role of ß1-integrin in CD cells, specifically in the PCs. We conditionally deleted ß1-integrin in mouse CD PCs using a specific aquaporin-2 (AQP2) promoter Cre-LoxP system. The resulting mutant mice, ß-1f/fAQP2-Cre+, had lower body weight, failed to thrive, and died around 8-12 wk. Their CD tubules were dilated, and some of them contained cellular debris. Increased apoptosis and proliferation of PCs were observed in the dilated CDs. Trichrome staining and electron microscopy revealed the presence of peritubular and interstitial fibrosis that is associated with increased production of extracellular matrix proteins including collagen type IV and fibronectin, as detected by immunoblotting. Further analysis revealed a significantly increased expression of transforming growth factor-ß (TGF-ß)-induced protein, fibronectin, and TGF-ß receptor-1 mRNAs and concomitantly increased phosphorylation of SMAD-2 that indicates the activation of the TGF-ß signaling pathway. Therefore, our data reveal that normal expression of ß1-integrin in PCs is a critical determinant of CD structural and functional integrity and further support the previously reported critical role of ß1-integrin in the development and/or maintenance of the CD structure and function.


Assuntos
Matriz Extracelular/metabolismo , Deleção de Genes , Integrina beta1/metabolismo , Medula Renal/metabolismo , Túbulos Renais Coletores/metabolismo , Poliúria/metabolismo , Insuficiência Renal/metabolismo , Fatores Etários , Animais , Apoptose , Aquaporina 2/genética , Proliferação de Células , Matriz Extracelular/ultraestrutura , Insuficiência de Crescimento/genética , Insuficiência de Crescimento/metabolismo , Insuficiência de Crescimento/patologia , Fibrose , Predisposição Genética para Doença , Integrases/genética , Integrina beta1/genética , Medula Renal/ultraestrutura , Túbulos Renais Coletores/ultraestrutura , Camundongos Knockout , Fenótipo , Fosforilação , Poliúria/genética , Poliúria/patologia , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Insuficiência Renal/genética , Insuficiência Renal/patologia , Transdução de Sinais , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/metabolismo
11.
Adv Exp Med Biol ; 969: 131-148, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28258571

RESUMO

Several aquaporin (AQP )-type water channels are expressed in kidney: AQP1 in the proximal tubule, thin descending limb of Henle, and vasa recta; AQP2 -6 in the collecting duct; AQP7 in the proximal tubule; AQP8 in the proximal tubule and collecting duct; and AQP11 in the endoplasmic reticulum of proximal tubule cells. AQP2 is the vasopressin-regulated water channel that is important in hereditary and acquired diseases affecting urine-concentrating ability. The roles of AQPs in renal physiology and transepithelial water transport have been determined using AQP knockout mouse models. This chapter describes renal physiologic insights revealed by phenotypic analysis of AQP knockout mice and the prospects for further basic and clinical studies.


Assuntos
Aquaporina 1/metabolismo , Rim/metabolismo , Poliúria/metabolismo , Ureia/metabolismo , Água/metabolismo , Animais , Aquaporina 1/genética , Transporte Biológico , Regulação da Expressão Gênica , Humanos , Rim/citologia , Capacidade de Concentração Renal/fisiologia , Camundongos , Camundongos Knockout , Concentração Osmolar , Poliúria/genética , Poliúria/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Equilíbrio Hidroeletrolítico/fisiologia
12.
Nat Commun ; 8: 14577, 2017 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-28233855

RESUMO

NF-E2-related factor-2 (Nrf2) regulates cellular responses to oxidative and electrophilic stress. Loss of Keap1 increases Nrf2 protein levels, and Keap1-null mice die of oesophageal hyperkeratosis because of Nrf2 hyperactivation. Here we show that deletion of oesophageal Nrf2 in Keap1-null mice allows survival until adulthood, but the animals develop polyuria with low osmolality and bilateral hydronephrosis. This phenotype is caused by defects in water reabsorption that are the result of reduced aquaporin 2 levels in the kidney. Renal tubular deletion of Keap1 promotes nephrogenic diabetes insipidus features, confirming that Nrf2 activation in developing tubular cells causes a water reabsorption defect. These findings suggest that Nrf2 activity should be tightly controlled during development in order to maintain renal homeostasis. In addition, tissue-specific ablation of Nrf2 in Keap1-null mice might create useful animal models to uncover novel physiological functions of Nrf2.


Assuntos
Diabetes Insípido Nefrogênico/patologia , Hidronefrose/patologia , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Fator 2 Relacionado a NF-E2/metabolismo , Poliúria/patologia , Animais , Aquaporina 2/metabolismo , Diferenciação Celular/genética , Diabetes Insípido Nefrogênico/genética , Modelos Animais de Doenças , Feminino , Humanos , Hidronefrose/genética , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 2 Relacionado a NF-E2/genética , Estresse Oxidativo/genética , Poliúria/genética , Reabsorção Renal/genética
13.
J Biol Chem ; 290(51): 30406-16, 2015 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-26453302

RESUMO

ClC-K chloride channels are crucial for auditory transduction and urine concentration. Mutations in CLCNKB, the gene encoding the renal chloride channel hClC-Kb, cause Bartter syndrome type III, a human genetic condition characterized by polyuria, hypokalemia, and alkalosis. In recent years, several Bartter syndrome-associated mutations have been described that result in truncations of the intracellular carboxyl terminus of hClC-Kb. We here used a combination of whole-cell patch clamp, confocal imaging, co-immunoprecipitation, and surface biotinylation to study the functional consequences of a frequent CLCNKB mutation that creates a premature stop codon at Trp-610. We found that W610X leaves the association of hClC-Kb and the accessory subunit barttin unaffected, but impairs its regulation by barttin. W610X attenuates hClC-Kb surface membrane insertion. Moreover, W610X results in hClC-Kb channel opening in the absence of barttin and prevents further barttin-mediated activation. To describe how the carboxyl terminus modifies the regulation by barttin we used V166E rClC-K1. V166E rClC-K1 is active without barttin and exhibits prominent, barttin-regulated voltage-dependent gating. Electrophysiological characterization of truncated V166E rClC-K1 demonstrated that the distal carboxyl terminus is necessary for slow cooperative gating. Since barttin modifies this particular gating process, channels lacking the distal carboxyl-terminal domain are no longer regulated by the accessory subunit. Our results demonstrate that the carboxyl terminus of hClC-Kb is not part of the binding site for barttin, but functionally modifies the interplay with barttin. The loss-of-activation of truncated hClC-Kb channels in heterologous expression systems fully explains the reduced basolateral chloride conductance in affected kidneys and the clinical symptoms of Bartter syndrome patients.


Assuntos
Canais de Cloreto/metabolismo , Ativação do Canal Iônico , Animais , Sítios de Ligação , Canais de Cloreto/genética , Códon sem Sentido/genética , Cães , Células HEK293 , Humanos , Hipopotassemia/genética , Hipopotassemia/metabolismo , Rim/metabolismo , Rim/patologia , Células Madin Darby de Rim Canino , Poliúria/genética , Poliúria/metabolismo , Poliúria/patologia , Transporte Proteico/genética
14.
Horm Res Paediatr ; 82(2): 138-42, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24819081

RESUMO

BACKGROUND: Familial hyperaldosteronism type III (FH-III) is a rare autosomal dominant disease for which five missense mutations in KCNJ5 have been identified. FH-III has a wide phenotypic variability from spironolactone-responsive hyperaldosteronism to massive adrenal hypertrophy with drug-resistant hypertension. This variation has mainly been attributed to genotype, because, in contrast to other genotypes (G151R, T158A, I157S, and Y152C), (1) FH-III patients with G151E have shown milder phenotype, and (2) G151E-harboring cells were found to have rapid lethality due to much larger sodium conductance of the encoded channel (Kir3.4), which prevents adrenal hypertrophy. METHODS: Here we describe the clinical course of a sporadic case of FH-III, with de novo G151R mutation. RESULTS: The patient developed polyuria at around 1.5 years of age and developed hypertension and hypokalemia by 4 years of age. Thereafter, spironolactone treatment successfully ameliorated hyperaldosteronism for 7 years with no discernible adrenal enlargement. CONCLUSION: Diverse clinical severity in FH-III cannot be defined solely by KCNJ5 genotype.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Mutação de Sentido Incorreto , Poliúria/genética , Glândulas Suprarrenais/patologia , Substituição de Aminoácidos , Pré-Escolar , Humanos , Hiperaldosteronismo/tratamento farmacológico , Hiperaldosteronismo/genética , Hiperaldosteronismo/patologia , Masculino , Antagonistas de Receptores de Mineralocorticoides/administração & dosagem , Poliúria/tratamento farmacológico , Poliúria/patologia , Espironolactona/administração & dosagem
15.
J Am Soc Nephrol ; 24(2): 243-52, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23308014

RESUMO

The mammalian collecting duct comprises principal and intercalated cells, which maintain sodium/water and acid/base balance, respectively, but the epigenetic contributors to the differentiation of these cell types remain unknown. Here, we investigated whether the histone H3 K79 methyltransferase Dot1l, which is highly expressed in principal cells, participates in this process. Taking advantage of the distribution of aquaporin 2 (Aqp2), which localizes to principal cells of the collecting duct, we developed mice lacking Dot1l in Aqp2-expressing cells (Dot1l(AC)) and found that these mice had approximately 20% fewer principal cells and 13%-16% more intercalated cells than control mice. This deletion of Dot1l in principal cells abolished histone H3 K79 methylation in these cells, but unexpectedly, most intercalated cells also had undetectable di-methyl K79, suggesting that Aqp2(+) cells give rise to intercalated cells. These Aqp2(+) cell-derived intercalated cells were present in both developing and mature kidneys. Furthermore, compared with control mice, Dot1l(AC) mice had 40% higher urine volume and 18% lower urine osmolarity with relatively normal electrolyte and acid-base homeostasis. In conclusion, these data suggest that Dot1l deletion facilitates the differentiation of some α- and ß-intercalated cells from Aqp2-expressing progenitor cells or mature principal cells.


Assuntos
Aquaporina 2/genética , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/fisiologia , Metiltransferases/genética , Equilíbrio Hidroeletrolítico/genética , Desequilíbrio Ácido-Base/genética , Desequilíbrio Ácido-Base/patologia , Desequilíbrio Ácido-Base/fisiopatologia , Animais , Aquaporina 2/metabolismo , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Epigênese Genética/fisiologia , Feminino , Histona-Lisina N-Metiltransferase , Histonas/metabolismo , Integrases/genética , Masculino , Metilação , Metiltransferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Poliúria/genética , Poliúria/patologia , Poliúria/fisiopatologia , Desequilíbrio Hidroeletrolítico/genética , Desequilíbrio Hidroeletrolítico/patologia , Desequilíbrio Hidroeletrolítico/fisiopatologia
16.
PLoS One ; 8(1): e53342, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23326416

RESUMO

Dot1l encodes histone H3 K79 methyltransferase Dot1a. Mice with Dot1l deficiency in renal Aqp2-expressing cells (Dot1l(AC)) develop polyuria by unknown mechanisms. Here, we report that Aqp5 links Dot1l deletion to polyuria through Aqp2. cDNA array analysis revealed and real-time RT-qPCR validated Aqp5 as the most upregulated gene in Dot1l(AC) vs. control mice. Aqp5 protein is barely detectable in controls, but robustly expressed in the Dot1l(AC) kidneys, where it colocalizes with Aqp2. The upregulation of Aqp5 is coupled with reduced association of Dot1a and H3 dimethyl K79 with specific subregions in Aqp5 5' flanking region in Dot1l(AC) vs. control mice. In vitro studies in IMCD3, MLE-15 and 293Tcells using multiple approaches including real-time RT-qPCR, luciferase reporter assay, cell surface biotinylation assay, colocalization, and co-immunoprecipitation uncovered that Dot1a represses Aqp5. Human AQP5 interacts with AQP2 and impairs its cell surface localization. The AQP5/AQP2 complex partially resides in the ER/Golgi. Consistently, AQP5 is expressed in none of 15 normal controls, but in all of 17 kidney biopsies from patients with diabetic nephropathy. In the patients with diabetic nephropathy, AQP5 colocalizes with AQP2 in the perinuclear region and AQP5 expression is associated with impaired cellular H3 dimethyl K79. Taken together, these data for the first time identify Aqp5 as a Dot1a potential transcriptional target, and an Aqp2 binding partner and regulator, and suggest that the upregulated Aqp5 may contribute to polyuria, possibly by impairing Aqp2 membrane localization, in Dot1l(AC) mice and in patients with diabetic nephropathy.


Assuntos
Aquaporina 2/metabolismo , Aquaporina 5/metabolismo , Metiltransferases/metabolismo , Transcrição Gênica , Sequência de Aminoácidos , Animais , Aquaporina 2/genética , Aquaporina 5/química , Aquaporina 5/genética , Sequência de Bases , Núcleo Celular/metabolismo , Imunoprecipitação da Cromatina , Nefropatias Diabéticas/complicações , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Histona-Lisina N-Metiltransferase , Histonas/metabolismo , Humanos , Rim/metabolismo , Rim/patologia , Lisina/metabolismo , Metilação , Camundongos , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Poliúria/complicações , Poliúria/genética , Poliúria/patologia , Ligação Proteica , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Regulação para Cima/genética
18.
Behav Brain Res ; 218(1): 106-13, 2011 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-21093493

RESUMO

Polydipsic hyponatremic schizophrenic (PHS) patients exhibit altered neuroendocrine activity that has been linked to their life-threatening water imbalance, as well as to impaired function and reduced volume of the anterior hippocampus. Polydipsic patients without hyponatremia (polydipsic normonatremic schizophrenics: PNS) exhibit similar, albeit less marked, changes in neuroendocrine activity and anterior hippocampal function, but not reduced anterior hippocampal volume. Indeed, reduced anterior hippocampal volume is seen in patients with normal water balance (nonpolydipsic normonatremic schizophrenics: NNS) whose neuroendocrine activity and anterior hippocampal function differ markedly from those with polydipsia. In an effort to reconcile these findings we measured hippocampal, amygdala and 3rd ventricle shapes in 26 schizophrenic patients (10 PNS, 7 PHS, 9 NNS) and 12 healthy controls matched for age and gender. Bilateral inward deformations were localized to the anterior lateral hippocampal surface (part of a neurocircuit which modulates neuroendocrine responses to psychological stimuli) in PHS and to a lesser extent in PNS, while deformations in NNS were restricted to the medial surface. Proportional deformations of the right medial amygdala, a key segment of this neurocircuit, were seen in both polydipsic groups, and correlated with the volume of the 3rd ventricle, which lies adjacent to the neuroendocrine nuclei. Finally, these structural findings were most marked in those with impaired hippocampal-mediated stress responses. These results reconcile previously conflicting data, and support the view that anterior lateral hippocampal pathology disrupts neuroendocrine function in polydipsic patients with and without hyponatremia. The relationship of these findings to the underlying mental illness remains to be established.


Assuntos
Tonsila do Cerebelo/patologia , Hipocampo/patologia , Hiponatremia/patologia , Poliúria/patologia , Esquizofrenia/patologia , Terceiro Ventrículo/patologia , Adulto , Tonsila do Cerebelo/fisiopatologia , Feminino , Hipocampo/fisiopatologia , Humanos , Hiponatremia/complicações , Hiponatremia/fisiopatologia , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Sistemas Neurossecretores/patologia , Sistemas Neurossecretores/fisiopatologia , Tamanho do Órgão , Poliúria/complicações , Poliúria/fisiopatologia , Esquizofrenia/complicações , Esquizofrenia/fisiopatologia , Terceiro Ventrículo/fisiopatologia , Sede/fisiologia
19.
J Neuroendocrinol ; 22(7): 754-7, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20492364

RESUMO

Familial neurohypophysial diabetes insipidus (FNDI), an autosomal dominant disorder, is mostly caused by mutations in the gene of neurophysin II (NPII), the carrier protein of arginine vasopressin (AVP). The analyses of knock-in mice expressing a mutant NPII that causes FNDI in humans demonstrated that polyuria progressed substantially in the absence of loss of AVP neurones. Morphological analyses revealed that inclusion bodies were present in the AVP neurones in the supraoptic nucleus and that the size and numbers of inclusion bodies gradually increased in parallel with the increases in urine volume. Electron microscopic analyses showed that aggregates existed in the endoplasmic reticulum (ER) of AVP neurones. These data suggest that cell death is not the primary cause of polyuria in FNDI, and that the aggregate formation in the ER is likely to be related to the pathogenesis of the progressive polyuria.


Assuntos
Diabetes Insípido Neurogênico/fisiopatologia , Poliúria/fisiopatologia , Animais , Arginina Vasopressina/genética , Arginina Vasopressina/metabolismo , Morte Celular , Diabetes Insípido Neurogênico/genética , Diabetes Insípido Neurogênico/patologia , Retículo Endoplasmático/patologia , Humanos , Corpos de Inclusão/patologia , Mutação , Neurônios/citologia , Neurônios/metabolismo , Neurônios/patologia , Neurofisinas/genética , Neurofisinas/metabolismo , Poliúria/genética , Poliúria/patologia , Núcleo Supraóptico/metabolismo , Núcleo Supraóptico/patologia
20.
Am J Physiol Renal Physiol ; 298(6): F1405-15, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20219826

RESUMO

The bumetanide-sensitive Na(+)-K(+)-2Cl(-) cotransporter NKCC2, located in the thick ascending limb of Henle's loop, plays a critical role in the kidney's ability to concentrate urine. In humans, loss-of-function mutations of the solute carrier family 12 member 1 gene (SLC12A1), coding for NKCC2, cause type I Bartter syndrome, which is characterized by prenatal onset of a severe polyuria, salt-wasting tubulopathy, and hyperreninemia. In this study, we describe a novel chemically induced, recessive mutant mouse line termed Slc12a1(I299F) exhibiting late-onset manifestation of type I Bartter syndrome. Homozygous mutant mice are viable and exhibit severe polyuria, metabolic alkalosis, marked increase in plasma urea but close to normal creatininemia, hypermagnesemia, hyperprostaglandinuria, hypotension,, and osteopenia. Fractional excretion of urea is markedly decreased. In addition, calcium and magnesium excretions are more than doubled compared with wild-type mice, while uric acid excretion is twofold lower. In contrast to hyperreninemia present in human disease, plasma renin concentration in homozygotes is not increased. The polyuria observed in homozygotes may be due to the combination of two additive factors, a decrease in activity of mutant NKCC2 and an increase in medullary blood flow, due to prostaglandin-induced vasodilation, that impairs countercurrent exchange of urea in the medulla. In conclusion, this novel viable mouse line with a missense Slc12a1 mutation exhibits most of the features of type I Bartter syndrome and may represent a new model for the study of this human disease.


Assuntos
Síndrome de Bartter/genética , Capacidade de Concentração Renal/genética , Rim/fisiopatologia , Mutação de Sentido Incorreto , Poliúria/genética , Simportadores de Cloreto de Sódio-Potássio/genética , Ureia/sangue , Aldeído Redutase/metabolismo , Sequência de Aminoácidos , Animais , Síndrome de Bartter/metabolismo , Síndrome de Bartter/patologia , Síndrome de Bartter/fisiopatologia , Biomarcadores/sangue , Pressão Sanguínea/genética , Peso Corporal , Densidade Óssea , Cálcio/sangue , Creatinina/sangue , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Fêmur/diagnóstico por imagem , Genótipo , Homozigoto , Rim/metabolismo , Rim/patologia , Magnésio/sangue , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Mutantes , Dados de Sequência Molecular , Mucoproteínas/metabolismo , Fenótipo , Poliúria/metabolismo , Poliúria/patologia , Poliúria/fisiopatologia , Radiografia , Renina/metabolismo , Índice de Gravidade de Doença , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto , Ácido Úrico/sangue , Uromodulina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA