Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Nanoscale ; 16(19): 9400-9405, 2024 May 16.
Article En | MEDLINE | ID: mdl-38651636

Tough carboxymethylcellulose nanofibers (CMF)/zirconium (Zr) hydrogels were easily obtained by a freeze-crosslinking method, where Zr-containing HCl solution was added to frozen CMF sol and the mixture was allowed to thaw. The Zr content of the hydrogels increased with increasing Zr concentration in the initial HCl solution. Furthermore, the mechanical strength increased with increasing Zr content. The Young's modulus value was improved by approximately 6 times compared to the CMF hydrogel without Zr, i.e., from 4.5 kPa to 27.2 kPa. The hydrogel had a porous structure with a pore size of 133 ± 37 µm and a CMF-Zr sheet structure around the pores. The obtained CMF-Zr hydrogel exhibited high adsorptivity for fluoride. The maximum adsorption capacity (Qmax) was estimated to be 24.1 mg g-1. This simple gelation method provides useful insights for the development of easy-to-handle hydrogel-based adsorbents.

2.
Carbohydr Polym ; 327: 121538, 2024 Mar 01.
Article En | MEDLINE | ID: mdl-38171650

We describe nanocellulose (NC) hydrogels formed from chemically unmodified NC by cellulose crystalline transformation and subsequent freeze cross-linking reaction. The freeze cross-linked NC hydrogel with macropores (~100 µm) was prepared by freezing a mixture of NC and NaOH (0.2 mol L-1), adding citric acid to the frozen mixture, and thawing it. Using NaOH and freezing together induced the crystalline transformation of NC from cellulose I to II via freeze concentration. After the crystalline transformation, cross-linking between the NC and CA in the freeze concentration layer provided a strong NC network structure, forming NC hydrogels with high mechanical strength. The structural changes in NC caused by NaOH, freezing, and freeze cross-linking on the angstrom to micrometer scale were investigated with FT-IR, SAXS, PXRD, and SEM. The freeze cross-linked NC hydrogel easily retained powder adsorbents in its inner space by mixing the NC-NaOH sol and the powder, and the hydrogel showed high removal efficiency for heavy metals. The results highlight the versatility of chemically unmodified celluloses in developing functional materials and suggest possible practical applications. This study also provides new insights into the efficient use of chemical reactions of cellulose under freezing conditions.

3.
Sci Rep ; 13(1): 19515, 2023 11 09.
Article En | MEDLINE | ID: mdl-37945736

Recent studies have shown that D-allose, a rare sugar, elicits antitumor effects on different types of solid cancers, such as hepatocellular carcinoma, non-small-cell lung cancer, and squamous cell carcinoma of the head and neck. In this study, we examined the effects of D-allose on the proliferation of human glioblastoma (GBM) cell lines (i.e., U251MG and U87MG) in vitro and in vivo and the underlying mechanisms. D-allose treatment inhibited the proliferation of U251MG and U87MG cells in a dose-dependent manner (3-50 mM). However, D-allose treatment did not affect cell cycles or apoptosis in these cells but significantly decreased the cell division frequency in both GBM cell lines. In a subcutaneous U87MG cell xenograft model, intraperitoneal injection of D-allose (100 mg/kg/day) significantly reduced the tumor volume in 28 days. These data indicate that D-allose-induced reduction in cell proliferation is associated with a subsequent decrease in the number of cell divisions, independent of cell-cycle arrest and apoptosis. Thus, D-allose could be an attractive additive to therapeutic strategies for GBM.


Carcinoma, Non-Small-Cell Lung , Glioblastoma , Liver Neoplasms , Lung Neoplasms , Humans , Glioblastoma/drug therapy , Cell Proliferation , Glucose/metabolism , Cell Division , Apoptosis , Cell Line, Tumor
4.
Sci Rep ; 13(1): 808, 2023 01 16.
Article En | MEDLINE | ID: mdl-36646875

Glioblastoma is characterized by a strong self-renewal potential and poor differentiated state. We have reported previously that the (pro)renin receptor [(P)RR] is a potential target for glioma therapy by silencing the (P)RR gene. Here, we have examined the effects of a monoclonal antibody against (P)RR on gliomagenesis. Human glioma cell lines (U251MG and U87MG) and a glioma stem cell line (MGG23) were used for the in vitro study. The expressions of the Wnt/ß-catenin signaling pathway (Wnt signaling pathway) components and stemness markers were measured by Western blotting. The effects of the (P)RR antibody on cell proliferation, sphere formation, apoptosis and migration were also examined. Subcutaneous xenografts were also examined in nude mice. Treatment with the (P)RR antibody reduced expression of Wnt signaling pathway components and stemness markers. Furthermore, the (P)RR antibody reduced cell proliferation and decreased sphere formation significantly. The treatment also suppressed migration and induced apoptosis. In a subcutaneous xenograft model, systemic administration of the (P)RR antibody reduced tumor volume significantly. These data show that treatment with the (P)RR antibody is a potential therapeutic strategy for treating glioblastoma.


Glioblastoma , Glioma , Mice , Animals , Humans , Prorenin Receptor , Glioblastoma/drug therapy , Glioblastoma/genetics , Antibodies, Monoclonal/metabolism , Mice, Nude , Cell Line, Tumor , Glioma/genetics , Wnt Signaling Pathway/genetics , Cell Proliferation , beta Catenin/metabolism , Gene Expression Regulation, Neoplastic
5.
Sci Rep ; 12(1): 5359, 2022 03 30.
Article En | MEDLINE | ID: mdl-35354900

Artificial intelligences are promising in future societies, and neural networks are typical technologies with the advantages such as self-organization, self-learning, parallel distributed computing, and fault tolerance, but their size and power consumption are large. Neuromorphic systems are biomimetic systems from the hardware level, with the same advantages as living brains, especially compact size, low power, and robust operation, but some well-known ones are non-optimized systems, so the above benefits are only partially gained, for example, machine learning is processed elsewhere to download fixed parameters. To solve these problems, we are researching neuromorphic systems from various viewpoints. In this study, a neuromorphic chip integrated with a large-scale integration circuit (LSI) and amorphous-metal-oxide semiconductor (AOS) thin-film synapse devices has been developed. The neuron elements are digital circuit, which are made in an LSI, and the synapse devices are analog devices, which are made of the AOS thin film and directly integrated on the LSI. This is the world's first hybrid chip where neuron elements and synapse devices of different functional semiconductors are integrated, and local autonomous learning is utilized, which becomes possible because the AOS thin film can be deposited without heat treatment and there is no damage to the underneath layer, and has all advantages of neuromorphic systems.


Semiconductors , Synapses , Biomimetics , Neural Networks, Computer , Oxides , Synapses/physiology
6.
Commun Biol ; 3(1): 724, 2020 11 27.
Article En | MEDLINE | ID: mdl-33247206

(Pro)renin receptor [(P)RR] has a role in various diseases, such as cardiovascular and renal disorders and cancer. Aberrant (P)RR expression is prevalent in pancreatic ductal adenocarcinoma (PDAC) which is the most common pancreatic cancer. Here we show whether aberrant expression of (P)RR directly leads to genomic instability in human pancreatic ductal epithelial (HPDE) cells. (P)RR-expressing HPDE cells show obvious cellular atypia. Whole genome sequencing reveals that aberrant (P)RR expression induces large numbers of point mutations and structural variations at the genome level. A (P)RR-expressing cell population exhibits tumour-forming ability, showing both atypical nuclei characterised by distinctive nuclear bodies and chromosomal abnormalities. (P)RR overexpression upregulates SWItch/Sucrose Non-Fermentable (SWI/SNF)-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a, member 5 (SMARCA5) through a direct molecular interaction, which results in the failure of several genomic stability pathways. These data reveal that aberrant (P)RR expression contributes to the early carcinogenesis of PDAC.


Adenosine Triphosphatases/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Pancreatic Neoplasms/metabolism , Receptors, Cell Surface/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Adenosine Triphosphatases/genetics , Animals , Carcinoma, Pancreatic Ductal/genetics , Cell Line , Cell Transformation, Neoplastic , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , Gene Expression Regulation, Neoplastic , Genomic Instability , Humans , Male , Mice , Mice, Inbred BALB C , Pancreatic Neoplasms/genetics , Up-Regulation , Whole Genome Sequencing
7.
Mol Cancer Ther ; 19(9): 1844-1855, 2020 09.
Article En | MEDLINE | ID: mdl-32669314

We previously reported that silencing of the PRR gene, which encodes the (pro)renin receptor [(P)RR], significantly reduced Wnt/ß-catenin-dependent development of pancreatic ductal adenocarcinoma (PDAC). Here, we examined the effects of a panel of blocking mAbs directed against the (P)RR extracellular domain on proliferation of the human PDAC cell lines PK-1 and PANC-1 in vitro and in vivo We observed that four rat anti-(P)RR mAbs induced accumulation of cells in the G0-G1-phase of the cell cycle and significantly reduced proliferation in vitro concomitant with an attenuation of Wnt/ß-catenin signaling. Systemic administration of the anti-(P)RR mAbs to nude mice bearing subcutaneous PK-1 xenografts significantly decreased tumor expression of active ß-catenin and the proliferation marker Ki-67, and reduced tumor growth. In contrast, treatment with the handle region peptide of (pro)renin did not inhibit tumor growth in vitro or in vivo, indicating that the effects of the anti-(P)RR mAbs were independent of the renin-angiotensin system. These data indicate that mAbs against human (P)RR can suppress PDAC cell proliferation by hindering activation of the Wnt/ß-catenin signaling pathway. Thus, mAb-mediated (P)RR blockade could be an attractive therapeutic strategy for PDAC.


Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Receptors, Cell Surface/chemistry , Vacuolar Proton-Translocating ATPases/chemistry , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Carcinoma, Pancreatic Ductal/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Male , Mice , Pancreatic Neoplasms/metabolism , Protein Domains , Rats , Receptors, Cell Surface/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays
8.
Br J Cancer ; 120(2): 229-237, 2019 01.
Article En | MEDLINE | ID: mdl-30555158

BACKGROUND: Although constitutive activating mutations in the Wnt/ß-catenin signalling pathway are important for colorectal cancer development, canonical signalling through Wnt ligands is essential for ß-catenin activation. Here, we investigated the role of (pro)renin receptor ((P)RR), a component of the Wnt receptor complex, in the pathogenesis of colorectal cancer. METHODS: (P)RR silencing was performed in human colorectal cancer cells containing constitutive activating mutations in the Wnt/ß-catenin pathway. (P)RR overexpression was induced in normal colon epithelial cells. Protein and mRNA levels of pathway components were detected, and Wnt signalling activity was measured using a ß-catenin reporter. Cell proliferative activity and apoptosis were evaluated using WST-1 assay and flow cytometry. Xenografts were induced in nude mice. RESULTS: (P)RR expression was greater in colorectal cancer tissues and cells than in normal colorectal samples. Patients with strong (P)RR expression took more proportion in groups with poorly-differentiated, advanced and rapidly-progressing cancers. (P)RR silencing attenuated the pathway in colorectal cancer cells, impaired their proliferation in vitro and vivo. (P)RR overexpression enhanced the pathway and proliferation of normal colon epithelial cells. CONCLUSIONS: Aberrant (P)RR expression promotes colorectal cancer through the Wnt/ß-catenin signalling pathway despite constitutive pathway-activating mutations. (P)RR is a potential diagnostic and therapeutic target for colorectal cancer.


Colorectal Neoplasms/genetics , Receptors, Cell Surface/genetics , Renin/genetics , Vacuolar Proton-Translocating ATPases/genetics , Wnt Signaling Pathway/genetics , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mutation , Xenograft Model Antitumor Assays , beta Catenin/genetics
9.
PLoS One ; 12(10): e0185600, 2017.
Article En | MEDLINE | ID: mdl-29053707

High glucose has been demonstrated to induce angiotensinogen (AGT) synthesis in the renal proximal tubular cells (RPTCs) of rats, which may further activate the intrarenal renin-angiotensin system (RAS) and contribute to diabetic nephropathy. This study aimed to investigate the effects of high glucose on AGT in the RPTCs of human origin and identify the glucose-responsive transcriptional factor(s) that bind(s) to the DNA sequences of AGT promoter in human RPTCs. Human kidney (HK)-2 cells were treated with normal glucose (5.5 mM) and high glucose (15.0 mM), respectively. Levels of AGT mRNA and AGT secretion of HK-2 cells were measured by real-time polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay (ELISA), respectively. Consecutive 5'-end deletion mutant constructs and different site-directed mutagenesis products of human AGT promoter sequences were respectively transfected into HK-2 cells, followed by AGT promoter activity measurement through dual luciferase assay. High glucose significantly augmented the levels of AGT mRNA and AGT secretion of HK-2 cells, compared with normal glucose treatment. High glucose also significantly augmented AGT promoter activity in HK-2 cells transfected with the constructs of human AGT promoter sequences, compared with normal glucose treatment. Hepatocyte nuclear factor (HNF)-5 was found to be one of the glucose-responsive transcriptional factors of AGT in human RPTCs, since the mutation of its binding sites within AGT promoter sequences abolished the above effects of high glucose on AGT promoter activity as well as levels of AGT mRNA and its secretion. The present study has demonstrated, for the first time, that high glucose augments AGT in human RPTCs through HNF-5, which provides a potential therapeutic target for diabetic nephropathy.


Angiotensinogen/metabolism , Forkhead Box Protein M1/metabolism , Glucose/administration & dosage , Kidney Tubules, Proximal/drug effects , Angiotensinogen/genetics , Animals , Cell Line , Enzyme-Linked Immunosorbent Assay , Glucose/pharmacology , Humans , Kidney Tubules, Proximal/metabolism , Promoter Regions, Genetic , RNA, Messenger/genetics , Rats , Transcription Factors/metabolism
10.
Exp Cell Res ; 358(2): 343-351, 2017 09 15.
Article En | MEDLINE | ID: mdl-28689812

Hypoxia predisposes renal fibrosis. This study was conducted to identify novel approaches to ameliorate the pathogenic effect of hypoxia. Using human proximal tubular epithelial cells we showed that a pan-phosphodiesterase (PDE) inhibitor, 3-isobutyl-1-methylxanthine (IBMX) dose and time dependently downregulated hypoxia-inducible factor 1α (HIF-1α) mRNA expression, which was further augmented by addition of a transcriptional inhibitor, actinomycin D. IBMX also increased the cellular cyclic adenosine monophosphate (cAMP) level. Luciferase assay showed that blocking of protein kinase A (PKA) using H89 reduced, while 8-Br-cAMP agonized the repression of HIF-1α promoter activity in hypoxic condition. Deletion of cAMP response element binding sites from the HIF-1α promoter abrogated the effect of IBMX. Western blot and immunofluorescent study confirmed that the CoCl2 induced increased HIF-1α protein in whole cell lysate and in nucleus was reduced by the IBMX. Through this process, IBMX attenuated both CoCl2 and hypoxia induced mRNA expressions of two pro-fibrogenic factors, platelet-derived growth factor B and lysyl oxidase. Moreover, IBMX reduced production of a mesenchymal transformation factor, ß-catenin; as well as protected against hypoxia induced cell-death. Taken together, our study showed novel evidence that the PDE inhibitor IBMX can downregulate the transcription of HIF-1α, and thus may attenuate hypoxia induced renal fibrosis.


1-Methyl-3-isobutylxanthine/pharmacology , Epithelial Cells/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Cell Hypoxia/drug effects , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Epithelial Cells/metabolism , Humans , Proto-Oncogene Proteins c-sis/metabolism , Signal Transduction/drug effects , Xanthines/pharmacology , beta Catenin/metabolism
11.
J Neurosurg ; 127(4): 819-828, 2017 Oct.
Article En | MEDLINE | ID: mdl-28059652

OBJECTIVE The (pro)renin receptor (PRR) plays an essential role in the early development of the central nervous system by activating the Wnt/ß-catenin signaling pathway. The authors investigated the potential role of the PRR in the pathogenesis of glioma. METHODS The authors performed immunohistochemical analysis to detect both the PRR and isocitrate dehydrogenase 1 with mutations involving arginine 132 ( IDH1R132H) in paraffin sections of 31 gliomas. Expression of the PRR and Wnt pathway components in cultured human glioma cell lines (U251MG, U87MG, and T98G) was measured using Western blotting. The effects of PRR short interfering RNA (siRNA) on glioma cell proliferation (WST-1 assay and direct cell counting) and apoptosis (flow cytometry and the caspase-3 assay) were also examined. RESULTS PRR expression was significantly higher in glioblastoma than in normal tissue or in lower grade glioma, regardless of IDH1R132H mutation. PRR expression was also higher in human glioblastoma cell lines than in human astrocytes. PRR expression showed a significant positive correlation with the Ki-67 labeling index, while it had a significant negative correlation with the survival time of glioma patients. Treatment with PRR siRNA significantly reduced expression of Wnt2, activated ß-catenin, and cyclin D1 by human glioblastoma cell lines, and it reduced the proliferative capacity of these cell lines and induced apoptosis. CONCLUSIONS This is the first evidence that the PRR has an important role in development of glioma by aberrant activation of the Wnt/ß-catenin signaling pathway. This receptor may be both a prognostic marker and a therapeutic target for glioma.


Brain Neoplasms/etiology , Glioma/etiology , Receptors, Cell Surface/physiology , Vacuolar Proton-Translocating ATPases/physiology , Wnt Signaling Pathway/physiology , Adult , Brain Neoplasms/chemistry , Female , Glioma/chemistry , Humans , Male , Middle Aged , Receptors, Cell Surface/analysis , Receptors, Cell Surface/biosynthesis , Tumor Cells, Cultured , Vacuolar Proton-Translocating ATPases/analysis , Vacuolar Proton-Translocating ATPases/biosynthesis
12.
Sci Rep ; 5: 8854, 2015 Mar 09.
Article En | MEDLINE | ID: mdl-25747895

Although Wnt/ß-catenin signaling is known to be aberrantly activated in PDAC, mutations of CTNNB1, APC or other pathway components are rare in this tumor type, suggesting alternative mechanisms for Wnt/ß-catenin activation. Recent studies have implicated the (pro)renin receptor ((P)RR) is related to the Wnt/ß-catenin signaling pathway. We therefore investigated the possible role of (P)RR in pancreatic carcinogenesis. Plasma s(P)RR levels were significantly (P < 0.0001) higher in patients with PDAC than in healthy matched controls. We also identified aberrant expression of (P)RR in premalignant PanIN and PDAC lesions and all the PDAC cell lines examined. Inhibiting (P)RR with an siRNA attenuated activation of Wnt/ß-catenin signaling pathway and reduced the proliferative ability of PDAC cells in vitro and the growth of engrafted tumors in vivo. Loss of (P)RR induced apoptosis of human PDAC cells. This is the first demonstration that (P)RR may be profoundly involved in ductal tumorigenesis in the pancreas.


Carcinogenesis/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Receptors, Cell Surface/blood , Vacuolar Proton-Translocating ATPases/blood , Wnt Signaling Pathway , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Humans , Male , Middle Aged
13.
J Agric Food Chem ; 61(23): 5558-64, 2013 Jun 12.
Article En | MEDLINE | ID: mdl-23683106

Black soybean seed coat has abundant levels of polyphenols such as anthocyanins (cyanidin 3-glucoside; C3G) and procyanidins (PCs). This study found that dietary black soybean seed coat extract (BE) ameliorates hyperglycemia and insulin sensitivity via the activation of AMP-activated protein kinase (AMPK) in type 2 diabetic mice. Dietary BE significantly reduced blood glucose levels and enhanced insulin sensitivity. AMPK was activated in the skeletal muscle and liver of diabetic mice fed BE. This activation was accompanied by the up-regulation of glucose transporter 4 in skeletal muscle and the down-regulation of gluconeogenesis in the liver. These changes resulted in improved hyperglycemia and insulin sensitivity in type 2 diabetic mice. In vitro studies using L6 myotubes showed that C3G and PCs significantly induced AMPK activation and enhanced glucose uptake into the cells.


AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus, Type 2/drug therapy , Glycine max/chemistry , Hyperglycemia/drug therapy , Insulin Resistance , Plant Extracts/administration & dosage , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/metabolism , Humans , Hyperglycemia/enzymology , Hyperglycemia/metabolism , Liver/drug effects , Liver/enzymology , Male , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Seeds/chemistry
14.
Front Biosci (Elite Ed) ; 5(2): 697-705, 2013 01 01.
Article En | MEDLINE | ID: mdl-23277024

Prorenin-induced intracellular signaling pathway is not fully elucidated. We investigated whether the (pro)renin receptor mediates epidermal growth factor (EGF) receptor transactivation through angiotensin (Ang) II-dependent and -independent pathways in human embryo kidney 293 cells. Prorenin (2 nmol/L) caused biphasic phosphorylation of EGF receptor (Tyr992) and extracellular signal-regulated kinase (ERK) 1/2, peaking at 5 minutes followed by a decrease and a second peak at 60-120 minutes, whereas EGF receptor (Tyr1068) and Src were phosphorylated at only 120 minutes. These prorenin-induced phosphorylation processes were inhibited by (pro)renin receptor siRNA. Similarly, Ang II type 1 (AT1) receptor blocker (ARB) or AT1 receptor siRNA completely inhibited prorenin-induced phosphorylation of EGF receptor (Tyr1068) and Src, as well as the late peaks of EGF receptor (Tyr992) and ERK 1/2. However, early peaks of EGF receptor (Tyr992) and ERK 1/2 at 5 minutes were not effectively blocked by ARB or AT1 receptor siRNA. Incubation with prorenin significantly increased Ang II levels of cell lysate. These data indicate that the (pro)renin receptor mediates EGF receptor transactivation in both Ang II-dependent and -independent pathways.


Angiotensin II/metabolism , ErbB Receptors/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Transcriptional Activation/physiology , Analysis of Variance , Blotting, Western , DNA Primers/genetics , ErbB Receptors/physiology , HEK293 Cells , Humans , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Receptors, Cell Surface/administration & dosage , Receptors, Cell Surface/genetics , Time Factors , Prorenin Receptor
15.
PLoS One ; 7(7): e41896, 2012.
Article En | MEDLINE | ID: mdl-22911865

BACKGROUND: Mineralocorticoid receptor (MR) antagonists attenuate renal injury in salt-sensitive hypertensive rats with low plasma aldosterone levels. We hypothesized that oxidative stress causes MR activation in high-salt-fed Dahl salt-sensitive rats. Furthermore, we determined if MR activation persisted and induced renal injury, even after switching from a high- to a normal-salt diet. METHODS AND FINDINGS: High-salt feeding for 4 weeks increased dihydroethidium fluorescence (DHE, an oxidant production marker), p22phox (a NADPH oxidase subunit) and serum and glucocorticoid-regulated kinase-1 (SGK1, an MR transcript) in glomeruli, compared with normal-salt feeding, and these changes persisted 4 weeks after salt withdrawal. Tempol treatment (0.5 mmol/L) during high-salt feeding abolished the changes in DHE fluorescence, p22phox and SGK1. Dietary salt reduction after a 4-week high-salt diet decreased both blood pressure and proteinuria, but was associated with significantly higher proteinuria than in normal control rats at 4 weeks after salt reduction. Administration of tempol during high-salt feeding, or eplerenone, an MR antagonist (100 mg/kg/day), started after salt reduction, recovered proteinuria to normal levels at 4 weeks after salt reduction. Paraquat, a reactive oxygen species generator, enhanced MR transcriptional activity in cultured rat mesangial cells and mouse podocytes. CONCLUSIONS: These results suggest that oxidative stress plays an important role in glomerular MR activation in Dahl salt-sensitive rats. Persistent MR activation even after reducing salt intake could limit the beneficial effects of salt restriction.


Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Oxidative Stress/drug effects , Receptors, Mineralocorticoid/metabolism , Sodium Chloride, Dietary/pharmacology , Adrenalectomy , Aldosterone/blood , Animals , Biomarkers/metabolism , Blood Pressure/drug effects , Cyclic N-Oxides/pharmacology , Ethidium/analogs & derivatives , Ethidium/metabolism , Feeding Behavior/drug effects , Genes, Reporter , Immediate-Early Proteins/metabolism , Kidney Glomerulus/enzymology , Kidney Glomerulus/physiopathology , Luciferases/metabolism , Male , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mice , Podocytes/drug effects , Podocytes/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteinuria/blood , Proteinuria/pathology , Proteinuria/physiopathology , Rats , Rats, Inbred Dahl , Sodium Chloride, Dietary/administration & dosage , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/metabolism , Spin Labels , Systole/drug effects , Time Factors
16.
J Hypertens ; 29(4): 696-705, 2011 Apr.
Article En | MEDLINE | ID: mdl-21252698

BACKGROUND: It is widely acknowledged that the (pro)renin receptor mediates angiotensin (Ang) II-dependent and Ang II-independent effects of prorenin. METHOD: We examined the effect of prorenin on vascular smooth muscle cell (VSMC) signal transduction, proliferation, and hypertrophy. RESULTS: Recombinant rat prorenin dose-dependently increased extracellular signal-regulated kinase (ERK) 1/2 and Akt phosphorylation in rat VSMCs. Prorenin also significantly increased cell number, and [H]-thymidine and [H]-leucine incorporation, which were attenuated by pretreatment with inhibitors for ERK kinase and phosphatidylinositol 3 kinase. Prorenin was also found to stimulate epidermal growth factor (EGF) receptor and Src phosphorylation. Pretreatment of VSMCs with an EGF receptor tyrosine kinase inhibitor and a Src inhibitor significantly attenuated the prorenin-induced increase in ERK 1/2 and Akt phosphorylation, as well as DNA and protein synthesis. Prorenin-induced phosphorylation of the EGF receptor, ERK 1/2, and Akt, as well as DNA and protein synthesis were all blocked by (pro)renin receptor siRNA, but not by an Ang II type 1 receptor blocker, candesartan, nor an Ang-converting enzyme inhibitor, captopril. CONCLUSION: These results reveal that prorenin directly stimulates VSMC proliferative and hypertrophic changes, dependent on the (pro)renin receptor, independent of Ang II. Furthermore, EGF receptor-mediated ERK 1/2 and Akt activation contributes to prorenin-dependent proliferative and hypertrophic effects in VSMCs.


ErbB Receptors/physiology , Muscle, Smooth, Vascular/physiology , Proto-Oncogene Proteins c-akt/metabolism , Renin/physiology , Animals , Cardiomegaly , Cell Proliferation , Cells, Cultured , Enzyme Activation , Rats , Signal Transduction
...