Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 6(8)2021 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-33724959

RESUMO

Nephrogenic diabetes insipidus (NDI) patients produce large amounts of dilute urine. NDI can be congenital, resulting from mutations in the type-2 vasopressin receptor (V2R), or acquired, resulting from medications such as lithium. There are no effective treatment options for NDI. Activation of PKA is disrupted in both congenital and acquired NDI, resulting in decreased aquaporin-2 phosphorylation and water reabsorption. We show that adenosine monophosphate-activated protein kinase (AMPK) also phosphorylates aquaporin-2. We identified an activator of AMPK, NDI-5033, and we tested its ability to increase urine concentration in animal models of NDI. NDI-5033 increased AMPK phosphorylation by 2.5-fold, confirming activation. It increased urine osmolality in tolvaptan-treated NDI rats by 30%-50% and in V2R-KO mice by 50%. Metformin, another AMPK activator, can cause hypoglycemia, which makes it a risky option for treating NDI patients, especially children. Rats with NDI receiving NDI-5033 showed no hypoglycemia in a calorie-restricted, exercise protocol. Congenital NDI therapy needs to be effective long-term. We administered NDI-5033 for 3 weeks and saw no reduction in efficacy. We conclude that NDI-5033 can improve urine concentration in animals with NDI and holds promise as a potential therapy for patients with congenital NDI due to V2R mutations.


Assuntos
Adenilato Quinase/efeitos dos fármacos , Diabetes Insípido Nefrogênico/metabolismo , Ativadores de Enzimas/farmacologia , Capacidade de Concentração Renal/efeitos dos fármacos , Adenilato Quinase/metabolismo , Animais , Aquaporina 2/metabolismo , Diabetes Insípido Nefrogênico/genética , Modelos Animais de Doenças , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Receptores de Vasopressinas/genética
2.
Cells ; 9(12)2020 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-33255239

RESUMO

Adrenomedullin (ADM) is a vasodilator that causes natriuresis and diuresis. However, the direct effect of ADM on osmotic water permeability in the rat inner medullary collecting duct (IMCD) has not been tested. We investigated whether ADM and its ADM receptor components (CRLR, RAMP2, and 3) are expressed in rat inner medulla (IM) and whether ADM regulates osmotic water permeability in isolated perfused rat IMCDs. The mRNAs of ADM, CRLR, and RAMP2 and 3 were detected in rat IM. Abundant protein of CRLR and RAMP3 were also seen but RAMP2 protein level was extremely low. Adding ADM (100 nM) to the bath significantly decreased osmotic water permeability. ADM significantly decreased aquaporin-2 (AQP2) phosphorylation at Serine 256 (pS256) and increased it at Serine 261 (pS261). ADM significantly increased cAMP levels in IM. However, inhibition of cAMP by SQ22536 further decreased ADM-attenuated osmotic water permeability. Stimulation of cAMP by roflumilast increased ADM-attenuated osmotic water permeability. Previous studies show that ADM also stimulates phospholipase C (PLC) pathways including protein kinase C (PKC) and cGMP. We tested whether PLC pathways regulate ADM-attenuated osmotic water permeability. Blockade of either PLC by U73122 or PKC by rottlerin significantly augmented the ADM-attenuated osmotic water permeability and promoted pS256-AQP2 but did change pS261-AQP2. Inhibition of cGMP by L-NAME did not change AQP2 phosphorylation. In conclusion, ADM primarily binds to the CRLR-RAMP3 receptor to initiate signaling pathways in the IM. ADM reduced water reabsorption through a PLC-pathway involving PKC. ADM-attenuated water reabsorption may be related to decreased trafficking of AQP2 to the plasma membrane. cAMP is not involved in ADM-attenuated osmotic water permeability.


Assuntos
Adrenomedulina/farmacologia , Medula Renal/efeitos dos fármacos , Osmose/efeitos dos fármacos , Permeabilidade/efeitos dos fármacos , Água/metabolismo , Aminopiridinas/farmacologia , Animais , Aquaporina 2/metabolismo , Benzamidas/farmacologia , Membrana Celular/efeitos dos fármacos , AMP Cíclico/metabolismo , Ciclopropanos/farmacologia , Fosforilação/efeitos dos fármacos , Proteína Quinase C/metabolismo , RNA Mensageiro/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos
3.
Hum Gene Ther ; 31(5-6): 367-375, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31950871

RESUMO

Renal fibrosis leads to end-stage renal disease, but antifibrotic drugs are difficult to develop. Chronic kidney disease often results in muscle wasting, and thereby increases morbidity and mortality. In this work, adeno-associated virus (AAV)-mediated overexpressing miR-29a was hypothesized to counteract renal fibrosis and muscle wasting through muscle-kidney crosstalk in unilateral ureteral obstruction (UUO) mice. miR-29a level was downregulated in the kidney and skeletal muscle of UUO mice. The secretion of exosome-encapsulated miR-29a increased in cultured skeletal muscle satellite cells and HEK293 renal cells after stimulation with serum from UUO mice. This result was confirmed by qPCR and microRNA deep sequencing in the serum exosomes of mice with obstructed ureters. A recombinant AAV-miR-29a was generated to overexpress miR-29a and injected into the tibialis anterior muscle of the mice 2 weeks before UUO surgery. AAV-miR-29a abrogated the UUO-induced upregulation of YY1 and myostatin in skeletal muscles. Renal fibrosis was also partially improved in the UUO mice with intramuscular AAV-miR-29a transduction. AAV-miR-29a overexpression reversed the increase in transforming growth factor ß, fibronectin, alpha-smooth muscle actin, and collagen 1A1 and 4A1 levels in the kidney of UUO mice. AAV-green fluorescent protein was applied to trace the AAV route in vivo, and fluorescence was significantly visible in the injected/uninjected muscles and in the kidneys. In conclusion, intramuscular AAV-miR-29a injection attenuates muscle wasting and ameliorates renal fibrosis by downregulating several fibrotic-related proteins in UUO mice.


Assuntos
Nefropatias/terapia , MicroRNAs/genética , MicroRNAs/uso terapêutico , Atrofia Muscular/terapia , Animais , Células Cultivadas , Dependovirus/genética , Modelos Animais de Doenças , Fibrose/genética , Fibrose/terapia , Terapia Genética , Vetores Genéticos , Células HEK293 , Humanos , Nefropatias/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atrofia Muscular/complicações , Células Satélites de Músculo Esquelético/metabolismo , Obstrução Ureteral/complicações
4.
FASEB J ; 33(12): 13590-13601, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31593640

RESUMO

Kidney fibrosis occurs in almost every type of chronic kidney disease. We found that microRNA (miR)-26a was decreased in the kidney, muscle, and exosomes of unilateral ureteral obstruction (UUO) mice. We hypothesized that exogenous miR-26 could suppresses renal fibrosis and muscle wasting in obstructive kidney disease. For this purpose, we generated exosomes that encapsulated miR-26, then injected these into skeletal muscle of UUO mice. The expression of miR-26a was elevated in serum exosomes from UUO mice following exosome-miR-26a injection. In these mice, muscle wasting has been ameliorated as evidenced by increased muscle weights. In addition, a muscle atrophy marker, myostatin, is increased in UUO muscle; provision of miR-26a abolished this increase. We detected a remote effect of exosomes containing miR-26a in UUO-induced renal fibrosis. The intervention of miR-26a attenuated UUO-induced renal fibrosis as determined by immunohistological assessment of α-smooth muscle actin and Masson's trichrome staining. Furthermore, exogenous miR-26a decreased the protein levels of 2 profibrosis proteins, connective tissue growth factor (CTGF) and TGF-ß1, in UUO kidney. Our data showed that exosomes containing miR-26a prevented muscle atrophy by inhibiting the transcription factor forkhead box O1. Likewise, the exosome-carried miR-26a limited renal fibrosis by directly suppressing CTGF. Our findings provide an experimental basis for exosome-mediated therapy of muscle atrophy and renal fibrosis.-Zhang, A., Wang, H., Wang, B., Yuan, Y., Klein, J. D., Wang, X. H. Exogenous miR-26a suppresses muscle wasting and renal fibrosis in obstructive kidney disease.


Assuntos
Exossomos/metabolismo , Fibrose/prevenção & controle , Regulação da Expressão Gênica , Nefropatias/prevenção & controle , MicroRNAs/administração & dosagem , Atrofia Muscular/prevenção & controle , Obstrução Ureteral/complicações , Animais , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Exossomos/genética , Fibrose/etiologia , Fibrose/patologia , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Nefropatias/etiologia , Nefropatias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Atrofia Muscular/etiologia , Atrofia Muscular/patologia
5.
Theranostics ; 9(7): 1864-1877, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31037144

RESUMO

Uremic cardiomyopathy and muscle atrophy are associated with insulin resistance and contribute to chronic kidney disease (CKD)-induced morbidity and mortality. We hypothesized that restoration of miR-26a levels would enhance exosome-mediated microRNA transfer to improve muscle wasting and cardiomyopathy that occur in CKD. Methods: Using next generation sequencing and qPCR, we found that CKD mice had a decreased level of miR-26a in heart and skeletal muscle. We engineered an exosome vector that contained Lamp2b, an exosomal membrane protein gene fused with a muscle-specific surface peptide that targets muscle delivery. We transfected this vector into muscle satellite cells and then transduced these cells with adenovirus that expresses miR-26a to produce exosomes encapsulated miR-26a (Exo/miR-26a). Exo/miR-26a was injected once per week for 8 weeks into the tibialis anterior (TA) muscle of 5/6 nephrectomized CKD mice. Results: Treatment with Exo/miR-26a resulted in increased expression of miR-26a in skeletal muscle and heart. Overexpression of miR-26a increased the skeletal muscle cross-sectional area, decreased the upregulation of FBXO32/atrogin-1 and TRIM63/MuRF1 and depressed cardiac fibrosis lesions. In the hearts of CKD mice, FoxO1 was activated, and connective tissue growth factor, fibronectin and collagen type I alpha 1 were increased. These responses were blunted by injection of Exo/miR-26a. Echocardiograms showed that cardiac function was improved in CKD mice treated with Exo/miR-26a. Conclusion: Overexpression of miR-26a in muscle prevented CKD-induced muscle wasting and attenuated cardiomyopathy via exosome-mediated miR-26a transfer. These results suggest possible therapeutic strategies for using exosome delivery of miR-26a to treat complications of CKD.


Assuntos
Exossomos/metabolismo , Fibrose/metabolismo , MicroRNAs/metabolismo , Atrofia Muscular/metabolismo , Miocárdio/metabolismo , Insuficiência Renal Crônica/metabolismo , Animais , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fibronectinas/metabolismo , Proteína Forkhead Box O1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia
6.
J Am Soc Nephrol ; 29(6): 1706-1719, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29773687

RESUMO

BackgroundNedd4-2 is an E3 ubiquitin-protein ligase that associates with transport proteins, causing their ubiquitylation, and then internalization and degradation. Previous research has suggested a correlation between Nedd4-2 and BP. In this study, we explored the effect of intercalated cell (IC) Nedd4-2 gene ablation on IC transporter abundance and function and on BP.Methods We generated IC Nedd4-2 knockout mice using Cre-lox technology and produced global pendrin/Nedd4-2 null mice by breeding global Nedd4-2 null (Nedd4-2-/- ) mice with global pendrin null (Slc26a4-/- ) mice. Mice ate a diet with 1%-4% NaCl; BP was measured by tail cuff and radiotelemetry. We measured transepithelial transport of Cl- and total CO2 and transepithelial voltage in cortical collecting ducts perfused in vitro Transporter abundance was detected with immunoblots, immunohistochemistry, and immunogold cytochemistry.Results IC Nedd4-2 gene ablation markedly increased electroneutral Cl-/HCO3- exchange in the cortical collecting duct, although benzamil-, thiazide-, and bafilomycin-sensitive ion flux changed very little. IC Nedd4-2 gene ablation did not increase the abundance of type B IC transporters, such as AE4 (Slc4a9), H+-ATPase, barttin, or the Na+-dependent Cl-/HCO3- exchanger (Slc4a8). However, IC Nedd4-2 gene ablation increased CIC-5 total protein abundance, apical plasma membrane pendrin abundance, and the ratio of pendrin expression on the apical membrane to the cytoplasm. IC Nedd4-2 gene ablation increased BP by approximately 10 mm Hg. Moreover, pendrin gene ablation eliminated the increase in BP observed in global Nedd4-2 knockout mice.Conclusions IC Nedd4-2 regulates Cl-/HCO3- exchange in ICs., Nedd4-2 gene ablation increases BP in part through its action in these cells.


Assuntos
Pressão Sanguínea/genética , Canais Epiteliais de Sódio/metabolismo , Transporte de Íons/genética , Ubiquitina-Proteína Ligases Nedd4/genética , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Amilorida/análogos & derivados , Amilorida/farmacologia , Animais , Bicarbonatos/metabolismo , Membrana Celular/metabolismo , Canais de Cloreto/metabolismo , Antiportadores de Cloreto-Bicarbonato/metabolismo , Cloretos/metabolismo , Troca Iônica , Túbulos Renais Coletores/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , ATPases Translocadoras de Prótons/metabolismo , Prótons , Reabsorção Renal/efeitos dos fármacos , Simportadores de Sódio-Bicarbonato/metabolismo , Transportadores de Sulfato/genética , Transportadores de Sulfato/metabolismo , Tiazidas/farmacologia
7.
J Cachexia Sarcopenia Muscle ; 9(4): 755-770, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29582582

RESUMO

BACKGROUND: The treatment of muscle wasting is accompanied by benefits in other organs, possibly resulting from muscle-organ crosstalk. However, how the muscle communicates with these organs is less understood. Two microRNAs (miRs), miR-23a and miR-27a, are located together in a gene cluster and regulate proteins that are involved in the atrophy process. MiR-23a/27a has been shown to reduce muscle wasting and act as an anti-fibrotic agent. We hypothesized that intramuscular injection of miR-23a/27a would counteract both muscle wasting and renal fibrosis lesions in a streptozotocin-induced diabetic model. METHODS: We generated an adeno-associated virus (AAV) that overexpresses the miR-23a∼27a∼24-2 precursor RNA and injected it into the tibialis anterior muscle of streptozotocin-induced diabetic mice. Muscle cross-section area (immunohistology plus software measurement) and muscle function (grip strength) were used to evaluate muscle atrophy. Fibrosis-related proteins were measured by western blot to monitor renal damage. In some cases, AAV-GFP was used to mimic the miR movement in vivo, allowing us to track organ redistribution by using the Xtreme Imaging System. RESULTS: The injection of AAV-miR-23a/27a increased the levels of miR-23a and miR-27a as well as increased phosphorylated Akt, attenuated the levels of FoxO1 and PTEN proteins, and reduced the abundance of TRIM63/MuRF1 and FBXO32/atrogin-1 in skeletal muscles. It also decreased myostatin mRNA and protein levels as well as the levels of phosphorylated pSMAD2/3. Provision of miR-23a/27a attenuates the diabetes-induced reduction of muscle cross-sectional area and muscle function. Curiously, the serum BUN of diabetic animals was reduced in mice undergoing the miR-23a/27a intervention. Renal fibrosis, evaluated by Masson trichromatic staining, was also decreased as were kidney levels of phosphorylated SMAD2/3, alpha smooth muscle actin, fibronectin, and collagen. In diabetic mice injected intramuscularly with AAV-GFP, GFP fluorescence levels in the kidneys showed linear correlation with the levels in injected muscle when examined by linear regression. Following intramuscular injection of AAV-miR-23a∼27a∼24-2, the levels of miR-23a and miR-27a in serum exosomes and kidney were significantly increased compared with samples from control virus-injected mice; however, no viral DNA was detected in the kidney. CONCLUSIONS: We conclude that overexpression of miR-23a/27a in muscle prevents diabetes-induced muscle cachexia and attenuates renal fibrosis lesions via muscle-kidney crosstalk. Further, this crosstalk involves movement of miR potentially through muscle originated exosomes and serum distribution without movement of AAV. These results could provide new approaches for developing therapeutic strategies for diabetic nephropathy with muscle wasting.


Assuntos
Regulação da Expressão Gênica , Rim/metabolismo , Rim/patologia , MicroRNAs/genética , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Animais , Dependovirus/genética , Modelos Animais de Doenças , Fibrose , Vetores Genéticos/genética , Camundongos , Modelos Biológicos , Imagem Molecular , Atrofia Muscular/patologia , Miostatina/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Transdução de Sinais , Transdução Genética
8.
J Am Soc Nephrol ; 29(4): 1097-1107, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29237738

RESUMO

Urinary concentrating ability is central to mammalian water balance and depends on a medullary osmotic gradient generated by a countercurrent multiplication mechanism. Medullary hyperosmolarity is protected from washout by countercurrent exchange and efficient removal of interstitial fluid resorbed from the loop of Henle and collecting ducts. In most tissues, lymphatic vessels drain excess interstitial fluid back to the venous circulation. However, the renal medulla is devoid of classic lymphatics. Studies have suggested that the fenestrated ascending vasa recta (AVRs) drain the interstitial fluid in this location, but this function has not been conclusively shown. We report that late gestational deletion of the angiopoietin receptor endothelial tyrosine kinase 2 (Tie2) or both angiopoietin-1 and angiopoietin-2 prevents AVR formation in mice. The absence of AVR associated with rapid accumulation of fluid and cysts in the medullary interstitium, loss of medullary vascular bundles, and decreased urine concentrating ability. In transgenic reporter mice with normal angiopoietin-Tie2 signaling, medullary AVR exhibited an unusual hybrid endothelial phenotype, expressing lymphatic markers (prospero homeobox protein 1 and vascular endothelial growth factor receptor 3) as well as blood endothelial markers (CD34, endomucin, platelet endothelial cell adhesion molecule 1, and plasmalemmal vesicle-associated protein). Taken together, our data redefine the AVRs as Tie2 signaling-dependent specialized hybrid vessels and provide genetic evidence of the critical role of AVR in the countercurrent exchange mechanism and the structural integrity of the renal medulla.


Assuntos
Angiopoietina-1/fisiologia , Angiopoietina-2/fisiologia , Líquido Extracelular/metabolismo , Capacidade de Concentração Renal/fisiologia , Medula Renal/irrigação sanguínea , Receptor TIE-2/fisiologia , Angiopoietina-1/deficiência , Angiopoietina-1/genética , Angiopoietina-2/deficiência , Angiopoietina-2/genética , Animais , Padronização Corporal , Linhagem da Célula , Endotélio Vascular , Genes Reporter , Idade Gestacional , Proteínas de Homeodomínio/análise , Doenças Renais Císticas/genética , Medula Renal/embriologia , Medula Renal/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miofibroblastos/patologia , Osmose , Receptor TIE-2/deficiência , Receptor TIE-2/genética , Circulação Renal , Transdução de Sinais , Proteínas Supressoras de Tumor/análise , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/análise
9.
Am J Physiol Renal Physiol ; 312(6): F1128-F1140, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28381463

RESUMO

Chronic kidney disease (CKD) causes loss of lean body mass by multiple mechanisms. This study examines whether autophagy-mediated proteolysis contributes to CKD-induced muscle wasting. We tested autophagy in the muscle of CKD mice with plantaris muscle overloading to mimic resistance exercise or with acupuncture plus low-frequency electrical stimulation (Acu/LFES) treatment. In CKD muscle, Bnip3, Beclin-1, and LC3II mRNAs and proteins were increased compared with those in control muscle, indicating autophagosome-lysosome formation induction. Acu/LFES suppressed the CKD-induced upregulation of autophagy. However, overloading increased autophagy-related proteins in normal and CKD muscle. Serum from uremic mice induces autophagy formation but did not increase the myosin degradation or actin break down in cultured muscle satellite cells. We examined mitochondrial biogenesis, copy number, and ATP production in cultured myotubes, and found all three aspects to be decreased by uremic serum. Inhibition of autophagy partially reversed this decline in cultured myotubes. In CKD mice, the mitochondrial copy number, biogenesis marker peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), mitochondrial transcription factor A (TFAM), and mitochondrial fusion marker Mitofusin-2 (Mfn2) are decreased. Both muscle overloading and Acu/LFES increased mitochondrial copy number, and reversed the CKD-induced decreases in PGC-1α, TFAM, and Mfn2. We conclude that the autophagy is activated in the muscle of CKD mice. However, myofibrillar protein is not directly broken down through autophagy. Instead, CKD-induced upregulation of autophagy leads to dysfunction of mitochondria and decrease of ATP production.


Assuntos
Autofagia , Mitocôndrias Musculares/patologia , Músculo Esquelético/patologia , Atrofia Muscular/etiologia , Insuficiência Renal Crônica/complicações , Trifosfato de Adenosina/metabolismo , Animais , Proteína Beclina-1/genética , Proteína Beclina-1/metabolismo , Linhagem Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Regulação da Expressão Gênica , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias Musculares/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Biogênese de Organelas , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Insuficiência Renal Crônica/sangue , Uremia/sangue
10.
Curr Opin Nephrol Hypertens ; 25(5): 444-51, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27367911

RESUMO

PURPOSE OF REVIEW: Urea is transported by urea transporter proteins in kidney, erythrocytes, and other tissues. Mice in which different urea transporters have been knocked out have urine-concentrating defects, which has led to the development and testing of urea transporters Slc14A2 (UT-A) and Slc14A1 (UT-B) inhibitors as urearetics. This review summarizes the knowledge gained during the past year on urea transporter regulation and investigations into the clinical potential of urearetics. RECENT FINDINGS: UT-A1 undergoes several posttranslational modifications that increase its function by increasing UT-A1 accumulation in the apical plasma membrane. UT-A1 is phosphorylated by protein kinase A, exchange protein activated by cyclic AMP, protein kinase Cα, and AMP-activated protein kinase, all at different serine residues. UT-A1 is also regulated by 14-3-3, which contributes to UT-A1 removal from the membrane. UT-A1 is glycosylated with various glycan moieties in animal models of diabetes mellitus. Transgenic expression of UT-A1 into UT-A1/UT-A3 knockout mice restores urine-concentrating ability. UT-B is present in descending vasa recta and urinary bladder, and is linked to bladder cancer. Inhibitors of UT-A and UT-B have been developed that result in diuresis with fewer abnormalities in serum electrolytes than conventional diuretics. SUMMARY: Urea transporters play critical roles in the urine-concentrating mechanism. Urea transport inhibitors are a promising new class of diuretic agent.


Assuntos
Transporte Biológico , Rim/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Ureia/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Diurese , Diuréticos/farmacologia , Glicosilação , Humanos , Proteínas de Membrana Transportadoras/genética , Fosforilação , Proteína Quinase C/metabolismo , Transportadores de Ureia
11.
Am J Physiol Renal Physiol ; 310(10): F1008-12, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26962099

RESUMO

Nephrogenic diabetes insipidus (NDI) is characterized by production of very large quantities of dilute urine due to an inability of the kidney to respond to vasopressin. Congenital NDI results from mutations in the type 2 vasopressin receptor (V2R) in ∼90% of families. These patients do not have mutations in aquaporin-2 (AQP2) or urea transporter UT-A1 (UT-A1). We tested adenosine monophosphate kinase (AMPK) since it is known to phosphorylate another vasopressin-sensitive transporter, NKCC2 (Na-K-2Cl cotransporter). We found AMPK expressed in rat inner medulla (IM). AMPK directly phosphorylated AQP2 and UT-A1 in vitro. Metformin, an AMPK activator, increased phosphorylation of both AQP2 and UT-A1 in rat inner medullary collecting ducts (IMCDs). Metformin increased the apical plasma membrane accumulation of AQP2, but not UT-A1, in rat IM. Metformin increased both osmotic water permeability and urea permeability in perfused rat terminal IMCDs. These findings suggest that metformin increases osmotic water permeability by increasing AQP2 accumulation in the apical plasma membrane but increases urea permeability by activating UT-A1 already present in the membrane. Lastly, metformin increased urine osmolality in mice lacking a V2R, a mouse model of congenital NDI. We conclude that AMPK activation by metformin mimics many of the mechanisms by which vasopressin increases urine-concentrating ability. These findings suggest that metformin may be a novel therapeutic option for congenital NDI due to V2R mutations.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Aquaporina 2/metabolismo , Diabetes Insípido Nefrogênico/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Proteínas de Membrana Transportadoras/metabolismo , Metformina/uso terapêutico , Proteínas Quinases Ativadas por AMP/efeitos dos fármacos , Animais , Diabetes Insípido Nefrogênico/urina , Avaliação Pré-Clínica de Medicamentos , Hipoglicemiantes/farmacologia , Metformina/farmacologia , Fosforilação/efeitos dos fármacos , Ratos Sprague-Dawley , Ureia/metabolismo , Água/metabolismo , Transportadores de Ureia
12.
J Appl Physiol (1985) ; 120(4): 426-36, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26679610

RESUMO

Muscle wasting occurs in a variety of clinical situations, including denervation. There is no effective pharmacological treatment for muscle wasting. In this study, we used a tibial nerve denervation model to test acupuncture plus low-frequency electric stimulation (Acu-LFES) as a therapeutic strategy for muscle atrophy. Acupuncture needles were connected to an SDZ-II electronic acupuncture device delivering pulses at 20 Hz and 1 mA; the treatment was 15 min daily for 2 wk. Acu-LFES prevented soleus and plantaris muscle weight loss and increased muscle cross-sectional area in denervated mice. The abundances of Pax7, MyoD, myogenin, and embryonic myosin heavy chain were significantly increased by Acu-LFES in both normal and denervated muscle. The number of central nuclei was increased in Acu-LFES-treated muscle fibers. Phosphorylation of Akt was downregulated by denervation leading to a decline in muscle mass; however, Acu-LFES prevented the denervation-induced decline largely by upregulation of the IGF-1 signaling pathway. Acu-LFES reduced the abundance of muscle catabolic proteins forkhead O transcription factor and myostatin, contributing to the attenuated muscle atrophy. Acu-LFES stimulated the expression of macrophage markers (F4/80, IL-1b, and arginase-1) and inflammatory cytokines (IL-6, IFNγ, and TNFα) in normal and denervated muscle. Acu-LFES also stimulated production of the muscle-specific microRNAs miR-1 and miR-206. We conclude that Acu-LFES is effective in counteracting denervation-induced skeletal muscle atrophy and increasing muscle regeneration. Upregulation of IGF-1, downregulation of myostatin, and alteration of microRNAs contribute to the attenuation of muscle atrophy in denervated mice.


Assuntos
Terapia por Acupuntura/métodos , Estimulação Elétrica/métodos , Denervação Muscular/efeitos adversos , Fibras Musculares Esqueléticas/metabolismo , Atrofia Muscular/terapia , Animais , Citocinas/metabolismo , Regulação para Baixo/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Proteínas Musculares/metabolismo , Atrofia Muscular/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Miostatina/metabolismo , Agulhas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Nervo Tibial/metabolismo , Regulação para Cima/fisiologia
13.
Am J Physiol Cell Physiol ; 309(9): C608-15, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26333598

RESUMO

Hypertonicity increases urea transport, as well as the phosphorylation and membrane accumulation of UT-A1, the transporter responsible for urea permeability in the inner medullary collect duct (IMCD). Hypertonicity stimulates urea transport through PKC-mediated phosphorylation. To determine whether PKC phosphorylates UT-A1, eight potential PKC phosphorylation sites were individually replaced with alanine and subsequently transfected into LLC-PK1 cells. Of the single mutants, only ablation of the S494 site dampened induction of total UT-A1 phosphorylation by the PKC activator phorbol dibutyrate (PDBu). This result was confirmed using a newly generated antibody that specifically detected phosphorylation of UT-A1 at S494. Hypertonicity increased UT-A1 phosphorylation at S494. In contrast, activators of cAMP pathways (PKA and Epac) did not increase UT-A1 phosphorylation at S494. Activation of both PKC and PKA pathways increased plasma membrane accumulation of UT-A1, although activation of PKC alone did not do so. However, ablating the PKC site S494 decreased UT-A1 abundance in the plasma membrane. This suggests that the cAMP pathway promotes UT-A1 trafficking to the apical membrane where the PKC pathway can phosphorylate the transporter, resulting in increased UT-A1 retention at the apical membrane. In summary, activation of PKC increases the phosphorylation of UT-A1 at a specific residue, S494. Although there is no cross talk with the cAMP-signaling pathway, phosphorylation of S494 through PKC may enhance vasopressin-stimulated urea permeability by retaining UT-A1 in the plasma membrane.


Assuntos
Membrana Celular/enzimologia , Medula Renal/enzimologia , Túbulos Renais Coletores/enzimologia , Proteínas de Membrana Transportadoras/metabolismo , Proteína Quinase C-alfa/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Soluções Hipertônicas/farmacologia , Medula Renal/efeitos dos fármacos , Túbulos Renais Coletores/efeitos dos fármacos , Células LLC-PK1 , Masculino , Proteínas de Membrana Transportadoras/genética , Camundongos , Mutagênese Sítio-Dirigida , Mutação , Osmose , Fosforilação , Proteína Quinase C-alfa/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico , Ratos Sprague-Dawley , Serina , Suínos , Transfecção , Transportadores de Ureia
14.
PLoS One ; 10(7): e0134511, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26230945

RESUMO

Mortality and morbidity are increased in patients with muscle atrophy resulting from catabolic diseases such as diabetes. At present there is no pharmacological treatment that successfully reverses muscle wasting from catabolic conditions. We hypothesized that acupuncture plus low frequency electric stimulation (Acu-LFES) would mimic the impact of exercise and prevent diabetes-induced muscle loss. Streptozotocin (STZ) was used to induce diabetes in mice. The mice were then treated with Acu-LFES for 15 minutes daily for 14 days. Acupuncture points were selected according to the WHO Standard Acupuncture Nomenclature guide. The needles were connected to an SDZ-II electronic acupuncture device delivering pulses at 20Hz and 1mA. Acu-LFES prevented soleus and EDL muscle weight loss and increased hind-limb muscle grip function in diabetic mice. Muscle regeneration capacity was significantly increased by Acu-LFES. The expression of Pax7, MyoD, myogenin and embryo myosin heavy chain (eMyHC) was significantly decreased in diabetic muscle vs. control muscle. The suppressed levels in diabetic muscle were reversed by Acu-LFES. The IGF-1 signaling pathway was also upregulated by Acu-LFES. Phosphorylation of Akt, mTOR and p70S6K were downregulated by diabetes leading to a decline in muscle mass, however, Acu-LFES countered the diabetes-induced decline. In addition, microRNA-1 and -206 were increased by Acu-LFES after 24 days of treatment. We conclude that Acu-LFES is effective in counteracting diabetes-induced skeletal muscle atrophy by increasing IGF-1 and its stimulation of muscle regeneration.


Assuntos
Complicações do Diabetes/terapia , Eletroacupuntura , Músculo Esquelético/fisiopatologia , Doenças Musculares/terapia , Regeneração , Animais , Complicações do Diabetes/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Proteínas Musculares/biossíntese , Doenças Musculares/etiologia , Doenças Musculares/fisiopatologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Estreptozocina
15.
Am J Physiol Renal Physiol ; 309(1): F71-8, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25995111

RESUMO

Urea transporter (UT)-A1 in the kidney inner medulla plays a critical role in the urinary concentrating mechanism and thereby in the regulation of water balance. The 14-3-3 proteins are a family of seven isoforms. They are multifunctional regulatory proteins that mainly bind to phosphorylated serine/threonine residues in target proteins. In the present study, we found that all seven 14-3-3 isoforms were detected in the kidney inner medulla. However, only the 14-3-3 γ-isoform was specifically and highly associated with UT-A1, as demonstrated by a glutathione-S-transferase-14-3-3 pulldown assay. The cAMP/adenylyl cyclase stimulator forskolin significantly enhanced their binding. Coinjection of 14-3-3γ cRNA into oocytes resulted in a decrease of UT-A1 function. In addition, 14-3-3γ increased UT-A1 ubiquitination and protein degradation. 14-3-3γ can interact with both UT-A1 and mouse double minute 2, the E3 ubiquitin ligase for UT-A1. Thus, activation of cAMP/PKA increases 14-3-3γ interactions with UT-A1 and stimulates mouse double minute 2-mediated UT-A1 ubiquitination and degradation, thereby forming a novel regulatory mechanism of urea transport activity.


Assuntos
Proteínas 14-3-3/metabolismo , Medula Renal/metabolismo , Rim/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Animais , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação para Baixo , Células Epiteliais/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Ratos Sprague-Dawley , Ubiquitinação , Transportadores de Ureia
16.
J Am Soc Nephrol ; 26(4): 926-34, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25300290

RESUMO

The urea transporter A1 (UT-A1) is a glycosylated protein with two glycoforms: 117 and 97 kD. In diabetes, the increased abundance of the heavily glycosylated 117-kD UT-A1 corresponds to an increase of kidney tubule urea permeability. We previously reported that diabetes not only causes an increase of UT-A1 protein abundance but also, results in UT-A1 glycan changes, including an increase of sialic acid content. Because activation of the diacylglycerol (DAG)-protein kinase C (PKC) pathway is elevated in diabetes and PKC-α regulates UT-A1 urea transport activity, we explored the role of PKC in UT-A1 glycan sialylation. We found that activation of PKC specifically promotes UT-A1 glycan sialylation in both UT-A1-MDCK cells and rat kidney inner medullary collecting duct suspensions, and inhibition of PKC activity blocks high glucose-induced UT-A1 sialylation. Overexpression of PKC-α promoted UT-A1 sialylation and membrane surface expression. Conversely, PKC-α-deficient mice had significantly less sialylated UT-A1 compared with wild-type mice. Furthermore, the effect of PKC-α-induced UT-A1 sialylation was mainly mediated by Src kinase but not Raf-1 kinase. Functionally, increased UT-A1 sialylation corresponded with enhanced urea transport activity. Thus, our results reveal a novel mechanism by which PKC regulates UT-A1 function by increasing glycan sialylation through Src kinase pathways, which may have an important role in preventing the osmotic diuresis caused by glucosuria under diabetic conditions.


Assuntos
Proteínas de Membrana Transportadoras/metabolismo , Proteína Quinase C-alfa/metabolismo , Ácidos Siálicos/metabolismo , Quinases da Família src/metabolismo , Animais , Cães , Células Madin Darby de Rim Canino , Masculino , Camundongos , Polissacarídeos/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Ratos Sprague-Dawley , Xenopus laevis , Transportadores de Ureia
17.
J Am Soc Nephrol ; 26(3): 626-35, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25228359

RESUMO

Effective therapeutic strategies to treat CKD-induced muscle atrophy are urgently needed. Low-frequency electrical stimulation (LFES) may be effective in preventing muscle atrophy, because LFES is an acupuncture technique that mimics resistance exercise by inducing muscle contraction. To test this hypothesis, we treated 5/6-nephrectomized mice (CKD mice) and control mice with LFES for 15 days. LFES prevented soleus and extensor digitorum longus muscle weight loss and loss of hind-limb muscle grip in CKD mice. LFES countered the CKD-induced decline in the IGF-1 signaling pathway and led to increases in markers of protein synthesis and myogenesis and improvement in muscle protein metabolism. In control mice, we observed an acute response phase immediately after LFES, during which the expression of inflammatory cytokines (IFN-γ and IL-6) increased. Expression of the M1 macrophage marker IL-1ß also increased acutely, but expression of the M2 marker arginase-1 increased 2 days after initiation of LFES, paralleling the change in IGF-1. In muscle cross-sections of LFES-treated mice, arginase-1 colocalized with IGF-1. Additionally, expression of microRNA-1 and -206, which inhibits IGF-1 translation, decreased in the acute response phase after LFES and increased at a later phase. We conclude that LFES ameliorates CKD-induced skeletal muscle atrophy by upregulation of the IGF-1 signaling pathway, which improves protein metabolism and promotes myogenesis. The upregulation of IGF-1 may be mediated by decreased expression of microRNA-1 and -206 and/or activation of M2 macrophages.


Assuntos
Terapia por Estimulação Elétrica , Fator de Crescimento Insulin-Like I/metabolismo , Proteínas Musculares/biossíntese , Atrofia Muscular/terapia , Insuficiência Renal Crônica/complicações , Animais , Macrófagos/fisiologia , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Desenvolvimento Muscular , Força Muscular , Músculo Esquelético/metabolismo , Distribuição Aleatória , Transdução de Sinais , Regulação para Cima
18.
Aging (Albany NY) ; 6(3): 160-75, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24659628

RESUMO

The mechanisms underlying the development of aging-induced muscle atrophy are unclear. By microRNA array and individual qPCR analyses, we found significant up-regulation of miR-29 in muscles of aged rodents vs. results in young. With aging, p85α, IGF-1 and B-myb muscle levels were lower while the expression of certain cell arrest proteins (p53, p16 and pRB) increased. When miR-29 was expressed in muscle progenitor cells (MPC), their proliferation was impaired while SA-ßgal expression increased signifying the development of senescence. Impaired MPC proliferation resulted from interactions between miR-29 and the 3'-UTR of p85a, IGF-1 and B-myb, suppressing the translation of these mediators of myoblast proliferation. In vivo, electroporation of miR-29 into muscles of young mice suppressed the proliferation and increased levels of cellular arrest proteins, recapitulating aging-induced responses in muscle. A potential stimulus of miR-29 expression is Wnt-3a since we found that exogenous Wnt-3a stimulated miR-29 expression 2.7-fold in primary cultures of MPCs. Thus, aging-induced muscle senescence results from activation of miR-29 by Wnt-3a leading to suppressed expression of several signaling proteins (p85α, IGF-1 and B-myb) that act coordinately to impair the proliferation of MPCs contributing to muscle atrophy. The increase in miR-29 provides a potential mechanism for aging-induced sarcopenia.


Assuntos
Envelhecimento/genética , Envelhecimento/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Sarcopenia/etiologia , Regiões 3' não Traduzidas , Envelhecimento/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Senescência Celular/genética , Senescência Celular/fisiologia , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/patologia , Regiões Promotoras Genéticas , Ratos , Ratos Endogâmicos F344 , Sarcopenia/genética , Sarcopenia/patologia , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação para Cima , Proteína Wnt3A/metabolismo
19.
Am J Physiol Cell Physiol ; 306(1): C28-36, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24196529

RESUMO

Using microarray analysis, we found that aging sarcopenia is associated with a sharp increase in the mRNA of the matricellular protein CCN1 (Cyr61/CTGF/Nov). CCN1 mRNA was upregulated 113-fold in muscle of aged vs. young rats. CCN1 protein was increased in aging muscle in both rats (2.8-fold) and mice (3.8-fold). When muscle progenitor cells (MPCs) were treated with recombinant CCN1, cell proliferation was decreased but there was no change in the myogenic marker myoD. However, the CCN1-treated MPCs did express a senescence marker (SA-ßgal). Interestingly, we found CCN1 increased p53, p16(Ink4A), and pRP (hypophosphorylated retinoblastoma protein) protein levels, all of which can arrest cell growth in MPCs. When MPCs were treated with aged rodent serum CCN1 mRNA increased by sevenfold and protein increased by threefold suggesting the presence of a circulating regulator. Therefore, we looked for a circulating regulator. Wnt-3a, a stimulator of CCN1 expression, was increased in serum from elderly humans (2.6-fold) and aged rodents (2.0-fold) compared with young controls. We transduced C2C12 myoblasts with wnt-3a and found that CCN1 protein was increased in a time- and dose-dependent manner. We conclude that in aging muscle, the circulating factor wnt-3a acts to increase CCN1 expression, prompting muscle senescence by activating cell arrest proteins.


Assuntos
Envelhecimento/metabolismo , Senescência Celular/fisiologia , Proteína Rica em Cisteína 61/biossíntese , Regulação da Expressão Gênica , Músculo Esquelético/metabolismo , Animais , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mioblastos Esqueléticos/metabolismo , RNA Mensageiro/biossíntese , Ratos , Ratos Endogâmicos F344 , Células-Tronco/metabolismo
20.
Biochim Biophys Acta ; 1833(10): 2143-52, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23684706

RESUMO

The mechanisms by which aldosterone increases Na(+), K(+) ATPase and sodium channel activity in cortical collecting duct and distal nephron have been extensively studied. Recent investigations demonstrate that aldosterone increases Na-H exchanger-3 (NHE-3) activity, bicarbonate transport, and H(+) ATPase in proximal tubules. However, the role of aldosterone in regulation of Na(+), K(+) ATPase in proximal tubules is unknown. We hypothesize that aldosterone increases Na(+), K(+) ATPase activity in proximal tubules through activation of the mineralocorticoid receptor (MR). Immunohistochemistry of kidney sections from human, rat, and mouse kidneys revealed that the MR is expressed in the cytosol of tubules staining positively for Lotus tetragonolobus agglutinin and type IIa sodium-phosphate cotransporter (NpT2a), confirming proximal tubule localization. Adrenalectomy in Sprague-Dawley rats decreased expression of MR, ENaC α, Na(+), K(+) ATPase α1, and NHE-1 in all tubules, while supplementation with aldosterone restored expression of above proteins. In human kidney proximal tubule (HKC11) cells, treatment with aldosterone resulted in translocation of MR to the nucleus and phosphorylation of SGK-1. Treatment with aldosterone also increased Na(+), K(+) ATPase-mediated (86)Rb uptake and expression of Na(+), K(+) ATPase α1 subunits in HKC11 cells. The effects of aldosterone on Na(+), K(+) ATPase-mediated (86)Rb uptake were prevented by spironolactone, a competitive inhibitor of aldosterone for the MR, and partially by Mifepristone, a glucocorticoid receptor (GR) inhibitor. These results suggest that aldosterone regulates Na(+), K(+) ATPase in renal proximal tubule cells through an MR-dependent mechanism.


Assuntos
Trifosfato de Adenosina/metabolismo , Aldosterona/farmacologia , Túbulos Renais Proximais/efeitos dos fármacos , Receptores de Mineralocorticoides/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Western Blotting , Membrana Celular , Células Cultivadas , Humanos , Hidrólise , Técnicas Imunoenzimáticas , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA