Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Basis Dis ; 1869(7): 166765, 2023 10.
Article in English | MEDLINE | ID: mdl-37245528

ABSTRACT

Lithium, mainstay treatment for bipolar disorder, frequently causes nephrogenic diabetes insipidus (NDI) and renal injury. However, the detailed mechanism remains unclear. Here we used the analysis of metabolomics and transcriptomics and metabolic intervention in a lithium-induced NDI model. Mice were treated with lithium chloride (40 mmol/kg chow) and rotenone (ROT, 100 ppm) in diet for 28 days. Transmission electron microscopy showed extensive mitochondrial structural abnormalities in whole nephron. ROT treatment markedly ameliorated lithium-induced NDI and mitochondrial structural abnormalities. Moreover, ROT attenuated the decrease of mitochondrial membrane potential in line with the upregulation of mitochondrial genes in kidney. Metabolomics and transcriptomics data demonstrated that lithium activated galactose metabolism, glycolysis, and amino sugar and nucleotide sugar metabolism. All these events were indicative of metabolic reprogramming in kidney cells. Importantly, ROT ameliorated metabolic reprogramming in NDI model. Based on transcriptomics analysis, we also found the activation of MAPK, mTOR and PI3K-Akt signaling pathways and impaired focal adhesion, ECM-receptor interaction and actin cytoskeleton in Li-NDI model were inhibited or attenuated by ROT treatment. Meanwhile, ROT administration inhibited the increase of Reactive Oxygen Species (ROS) in NDI kidneys along with enhanced SOD2 expression. Finally, we observed that ROT partially restored the reduced AQP2 and enhanced urinary sodium excretion along with the blockade of increased PGE2 output. Taken together, the current study demonstrates that mitochondrial abnormalities and metabolic reprogramming play a key role in lithium-induced NDI, as well as the dysregulated signaling pathways, thereby serving as a novel therapeutic target.


Subject(s)
Diabetes Insipidus, Nephrogenic , Diabetes Mellitus , Mice , Animals , Diabetes Insipidus, Nephrogenic/chemically induced , Diabetes Insipidus, Nephrogenic/genetics , Diabetes Insipidus, Nephrogenic/metabolism , Lithium/pharmacology , Aquaporin 2/genetics , Aquaporin 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Kidney/metabolism
2.
Oncogene ; 40(47): 6479-6493, 2021 11.
Article in English | MEDLINE | ID: mdl-34611310

ABSTRACT

Androgen receptor (AR) plays a central role in driving prostate cancer (PCa) progression. How AR promotes this process is still not completely clear. Herein, we used single-cell transcriptome analysis to reconstruct the transcriptional network of AR in PCa. Our work shows AR directly regulates a set of signature genes in the ER-to-Golgi protein vesicle-mediated transport pathway. The expression of these genes is required for maximum androgen-dependent ER-to-Golgi trafficking, cell growth, and survival. Our analyses also reveal the signature genes are associated with PCa progression and prognosis. Moreover, we find inhibition of the ER-to-Golgi transport process with a small molecule enhanced antiandrogen-mediated tumor suppression of hormone-sensitive and insensitive PCa. Finally, we demonstrate AR collaborates with CREB3L2 in mediating ER-to-Golgi trafficking in PCa. In summary, our findings uncover a critical role for dysregulation of ER-to-Golgi trafficking expression and function in PCa progression, provide detailed mechanistic insights for how AR tightly controls this process, and highlight the prospect of targeting the ER-to-Golgi pathway as a therapeutic strategy for advanced PCa.


Subject(s)
Androgens/pharmacology , Basic-Leucine Zipper Transcription Factors/metabolism , Endoplasmic Reticulum/pathology , Gene Expression Regulation, Neoplastic/drug effects , Golgi Apparatus/pathology , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Animals , Apoptosis , Basic-Leucine Zipper Transcription Factors/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Gene Regulatory Networks , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Humans , Male , Mice , Prognosis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Single-Cell Analysis/methods , Survival Rate , Transcriptome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Front Physiol ; 12: 642274, 2021.
Article in English | MEDLINE | ID: mdl-33868005

ABSTRACT

Emerging evidence is showing that apelin plays an important role in regulating salt and water balance by counteracting the antidiuretic action of vasopressin (AVP). However, the underlying mechanism remains unknown. Here, we hypothesized that (pro) renin receptor (PRR)/soluble prorenin receptor (sPRR) might mediate the diuretic action of apelin in the distal nephron. During water deprivation (WD), the urine concentrating capability was impaired by an apelin peptide, apelin-13, accompanied by the suppression of the protein expression of aquaporin 2 (AQP2), NKCC2, PRR/sPRR, renin and nuclear ß-catenin levels in the kidney. The upregulated expression of AQP2 or PRR/sPRR both induced by AVP and 8-Br-cAMP was blocked by apelin-13, PKA inhibitor (H89), or ß-catenin inhibitor (ICG001). Interestingly, the blockage of apelin-13 on AVP-induced AQP2 protein expression was reversed by exogenous sPRR. Together, the present study has defined the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA)/sPRR pathway in the CD as the molecular target of the diuretic action of apelin.

4.
Hypertens Res ; 44(2): 168-178, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32908237

ABSTRACT

Our recent work demonstrates that infusion of sodium butyrate (NaBu) into the renal medulla blunts angiotensin II-induced hypertension and improves renal injury. The present study aimed to test whether oral administration of NaBu attenuates salt-sensitive hypertension in deoxycorticosterone acetate (DOCA)/salt-treated rats. Uninephrectomized male Sprague-Dawley (SD) rats were treated with DOCA pellets (150 mg/rat) plus 1% NaCl drinking water for 2 weeks. Animals received oral administration of NaBu (1 g/kg) or vehicle once per day. Our results showed that NaBu administration significantly attenuated DOCA/salt-increased mean arterial pressure from 156 ± 4 mmHg to 136 ± 1 mmHg. DOCA/salt treatment markedly enhanced renal damage as indicated by an increased ratio of kidney weight/body weight, elevated urinary albumin, extensive fibrosis, and inflammation, whereas kidneys from NaBu-treated rats exhibited a significant reduction in these renal damage responses. Compared to the DOCA/salt group, the DOCA/salt-NaBu group had ~30% less salt water intake and decreased Na+ and Cl- excretion in urine but no alteration in 24-h urine excretion. Mechanistically, NaBu inhibited the protein levels of several sodium transporters stimulated by DOCA/salt in vivo, such as ßENaC, γENaC, NCC, and NKCC-2. Further examination showed that NaBu downregulated the expression of mineralocorticoid receptor (MR) and serum and glucocorticoid-dependent protein kinase 1 (SGK1) in DOCA/salt-treated rats or aldosterone-treated human renal tubular duct epithelial cells. These results provide evidence that NaBu may attenuate DOCA/salt-induced hypertension and renal damage by inhibiting the MR/SGK1 pathway.


Subject(s)
Hypertension , Kidney Diseases , Acetates , Animals , Blood Pressure , Butyric Acid , Desoxycorticosterone Acetate , Hypertension/chemically induced , Hypertension/complications , Hypertension/drug therapy , Immediate-Early Proteins , Kidney , Kidney Diseases/etiology , Male , Protein Serine-Threonine Kinases , Rats , Rats, Sprague-Dawley , Receptors, Mineralocorticoid , Signal Transduction , Sodium , Sodium Chloride , Sodium Chloride, Dietary
6.
Nutrition ; 75-76: 110766, 2020.
Article in English | MEDLINE | ID: mdl-32305658

ABSTRACT

OBJECTIVES: Although it is known that high fructose intake causes salt-sensitive hypertension, the underlying mechanism remains unclear. The aim of this study was to determine whether chronic intake of high fructose coupled with salt (HFS) might alter the structure of the gut microbiota, which contributes to elevated blood pressure. METHODS: For 8 wk, Sprague-Dawley rats were given 20% fructose in drinking water and 4% sodium chloride in their diet to induce hypertension. A non-absorbable antibiotic vancomycin was used to modify gut microbiota. The 16 S rRNA sequencing for fecal samples was assessed and blood pressure was recorded. Enzyme-linked immunosorbent assay and quantitative polymerase chain reaction were used to examine the renin-angiotensin system in serum, urine, and the kidney. RESULTS: Compared with the control group, HFS feeding resulted in gut dysbiosis by altering the diversity and richness of gut microbiota and decreased the ratio of Firmicutes to Bacteroidetes. Vancomycin reshaped dramatically the HFS-induced dysbiosis. And vancomycin (van) attenuated HFS-increased blood pressure (HFS: 121.3 ± 2.8 mm Hg; HFS-van: 111.1 ± 1.7 mm Hg) and heart rate (HFS: 360.5 ± 9.0 bpm; HFS-van: 318.7 ± 5.6 bpm) as well as the content of angiotensinogen, renin, and angiotensin II in the urine and the angiotensinogen mRNA level in renal cortical tissues. However, HFS-increased triacylglycerol, renin, and angiotensin II in serum were not decreased by vancomycin. CONCLUSION: The present results demonstrated that gut dysbiosis develops after chronic fructose plus salt intake and contributes to the increase of blood pressure and the activation of the intrarenal renin-angiotensin system. Therefore, targeting gut microbiota provides a helpful therapy method to improve HFS-induced hypertension.


Subject(s)
Hypertension , Sodium Chloride, Dietary , Animals , Blood Pressure , Dysbiosis/chemically induced , Dysbiosis/metabolism , Fructose/adverse effects , Hypertension/chemically induced , Kidney/metabolism , Rats , Rats, Sprague-Dawley , Renin-Angiotensin System , Sodium Chloride/metabolism , Sodium Chloride, Dietary/adverse effects
7.
J Cell Mol Med ; 23(12): 8139-8150, 2019 12.
Article in English | MEDLINE | ID: mdl-31565858

ABSTRACT

Sodium butyrate (NaBu) is reported to play important roles in a number of chronic diseases. The present work is aimed to investigate the effect of NaBu on angiotensin II (Ang II)-induced cardiac hypertrophy and the underlying mechanism in in vivo and in vitro models. Sprague Dawley rats were infused with vehicle or Ang II (200 ng/kg/min) and orally administrated with or without NaBu (1 g/kg/d) for two weeks. Cardiac hypertrophy parameters and COX2/PGE2 pathway were analysed by real-time PCR, ELISA, immunostaining and Western blot. The cardiomyocytes H9C2 cells were used as in vitro model to investigate the role of NaBu (2 mmol/L) in inhibition of Ang II-induced cardiac hypertrophy. NaBu significantly attenuated Ang II-induced increase in the mean arterial pressure. Ang II treatment remarkably increased cardiac hypertrophy as indicated by increased ratio of heart weight/body weight and enlarged cardiomyocyte size, extensive fibrosis and inflammation, as well as enhanced expression of hypertrophic markers, whereas hearts from NaBu-treated rats exhibited a significant reduction in these hypertrophic responses. Mechanistically, NaBu inhibited the expression of COX2/PGE2 along with production of ANP and phosphorylated ERK (pERK) stimulated by Ang II in in vivo and in vitro, which was accompanied by the suppression of HDAC5 and HDAC6 activities. Additionally, knocking down the expression of HDAC5 and HDAC6 via gene-editing strategy dramatically blocked Ang II-induced hypertrophic responses through COX2/PGE2 pathway. These results provide solid evidence that NaBu attenuates Ang II-induced cardiac hypertrophy by inhibiting the activation of COX2/PGE2 pathway in a HDAC5/HDAC6-dependent manner.


Subject(s)
Butyric Acid/pharmacology , Cardiomegaly/prevention & control , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Histone Deacetylase 6/metabolism , Histone Deacetylases/metabolism , Angiotensin II , Animals , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Cell Line , Cyclooxygenase 2/genetics , Gene Expression/drug effects , Gene Knockdown Techniques , Histone Deacetylase 6/genetics , Histone Deacetylases/genetics , Hypertension/chemically induced , Hypertension/genetics , Hypertension/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/genetics
8.
Am J Physiol Renal Physiol ; 316(3): F530-F538, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30379098

ABSTRACT

The (pro)renin receptor (PRR) is a new component of the renin-angiotensin-aldosterone system (RAAS) and regulates renin activity. The objective of the present study was to test potential roles of the renal PRR and intrarenal RAAS in the physiological status of late pregnancy. Late pregnant Sprague-Dawley rats were studied 19-21 days after sperm was observed in vaginal smears. Experiments were performed using age-matched virgin rats and late pregnant rats treated with the specific PRR inhibitor PRO20 (700 µg·kg-1·day-1 sc for 14 days, 3 times/day for every 8 h) or vehicle. The indices of RAAS, including PRR, renin, angiotensin II, and aldosterone levels, were examined by immunoblotting, qRT-PCR, or ELISA. Further analyses of renal epithelial sodium channel (ENaC) expression, sodium-water retention, and plasma volume were performed. We first present evidence for the activation of intrarenal RAAS in late pregnant rats, including increases in urinary renin activity, active and total renin content, and prorenin content, angiotensin II and aldosterone excretion, in parallel with increased renal PRR expression and urinary soluble PRR excretion. Functional evidence demonstrated that PRR antagonism with PRO20 effectively suppressed the indices of intrarenal RAAS in late pregnant rats. In addition, our results revealed that renal α-ENaC expression, sodium-water retention, and plasma volume were elevated during late pregnancy, which were all attenuated by PRO20. In summary, the present study examined the renal mechanism of sodium-water retention and plasma volume expansion in late pregnant rats and identified a novel role of PRR in regulation of intrarenal RAAS and α-ENaC and thus sodium and fluid retention associated with pregnancy.


Subject(s)
Epithelial Sodium Channels/metabolism , Kidney/metabolism , Receptors, Cell Surface/metabolism , Renin-Angiotensin System/physiology , Water-Electrolyte Imbalance/metabolism , Aldosterone/metabolism , Angiotensin II/metabolism , Animals , Blood Pressure/drug effects , Epithelial Sodium Channels/genetics , Female , Kidney/drug effects , Peptide Fragments/pharmacology , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics , Renin/pharmacology , Renin-Angiotensin System/drug effects , Sodium/metabolism , Water/metabolism , Prorenin Receptor
9.
Front Physiol ; 10: 1565, 2019.
Article in English | MEDLINE | ID: mdl-32038274

ABSTRACT

BACKGROUND: High salt intake is associated with both oxidative stress and chronic kidney disease (CKD) progression. Nuclear factor E2-related factor 2 (Nrf2) is a transcriptional factor regulating the antioxidant and detoxifying genes to potently antagonize oxidative stress. This study examined the effect of high salt loading on the expression of Nrf2 in kidney. METHODS: Mice were treated with acute salt loading, and Nrf2 expression in the kidney was detected by Western blotting and immunostaining. Reactive oxygen species (ROS) levels in the kidney were measured using dihydroethidium (DHE) staining. In vitro, mpkCCD cells were cultured in high osmolality medium by adding sodium chloride (NaCl), sodium gluconate (Na-Glu), choline chloride (Choline-Cl), or mannitol. Then, Nrf2 and its target genes were measured. RESULTS: Nrf2 protein in renal cortex and medulla tissue lysates was significantly downregulated after acute salt loading. Immunofluorescence data showed that Nrf2 was mainly located in collecting duct principal cells evidenced by co-staining of Nrf2 with AQP2. Contrasting to the reduced Nrf2 expression, ROS levels in the kidney were significantly increased after salt loading. In vitro, the Nrf2 protein level was downregulated in mpkCCD cells after NaCl treatment for 24 h. Interestingly, sodium gluconate had a similar effect on downregulating Nrf2 expression as NaCl, whereas neither Choline-Cl nor mannitol changed Nrf2 expression. Meanwhile, the mRNA levels of Nrf2 target genes were downregulated by NaCl and/or sodium gluconate, while some of them were also regulated by Choline-Cl, indicating a more complex regulation of these genes under a high salt condition. Finally, we found that the downregulation of Nrf2 caused by NaCl was not affected by N-acetylcysteine (NAC), spironolactone, or NS-398, suggesting other mechanisms mediating Nrf2 downregulation caused by high salt challenge. CONCLUSION: High salt downregulated Nrf2 mainly via a sodium-dependent manner in kidney collecting duct cells, which might contribute to the excessive renal oxidative stress and CKD progression.

10.
Physiol Rep ; 6(13): e13630, 2018 07.
Article in English | MEDLINE | ID: mdl-29998554

ABSTRACT

Hydrogen sulfide (H2 S) is recognized as a novel gasotransmitter involved in the regulation of nervous system, cardiovascular functions, inflammatory response, gastrointestinal system, and renal function. Cystathionine ß-synthase (CBS) and cystathionine γ-lyase (CSE) are the major enzymes responsible for H2 S production through desulfuration reactions. H2 S is reported to play a protective role in both high-fat diet (HFD)-induced obese and diabetic mice. However, the synthesizing enzyme involved is not clearly elucidated. The current study was aimed to investigate the regulation of CBS and CSE in different tissues including the kidney, liver, and epididymal fat in C57BL/6 mice after a HFD (60% kcal fat) for 24 weeks. The protein and mRNA expression of CBS was specifically decreased in the kidney while CSE remained unchanged, which was further confirmed in db/db mice. In the liver, CSE expression was downregulated after HFD accompanied with unchanged CBS. Moreover, CSE expression was even upregulated in epididymal fat. The specific downregulation of renal CBS may contribute to decreased H2 S production, which could be a pathogenic mechanism of obesity. Increased CSE/H2 S pathway in epididymal fat possibly resulted in impaired glucose uptake and aggravated insulin resistance. In conclusion, our results revealed that CBS was selectively downregulated in both diet and gene-induced obesity models.


Subject(s)
Cystathionine beta-Synthase/metabolism , Kidney/metabolism , Obesity/metabolism , Adipose Tissue/metabolism , Animals , Cystathionine beta-Synthase/genetics , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Diet, High-Fat/adverse effects , Hydrogen Sulfide/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/etiology
11.
Am J Physiol Renal Physiol ; 315(6): F1759-F1768, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29846109

ABSTRACT

Proteinuria is not only a common feature of chronic kidney diseases (CKD) but also an independent risk factor promoting CKD progression to end-stage renal failure. However, the underlying molecular mechanisms for protein overload-induced renal injury remain elusive. The present study examined the role of (pro)renin receptor (PRR) in pathogenesis of albumin overload (AO)-induced nephropathy and activation of the intrarenal renin-angiotensin system (RAS) in rats. Wistar rats underwent unilateral nephrectomy and were treated for 7 wk with vehicle, bovine serum albumin (5 g·kg-1·day-1 via a single ip injection), alone or in conjunction with the PRR decoy inhibitor PRO20 (500 µg·kg-1·day-1 via 3 sc injections). The AO rat model exhibited severe proteinuria, tubular necrosis, and interstitial fibrosis, oxidative stress, and inflammation, accompanied by elevated urinary N-acetyl-ß-d-glucosaminidase activity and urinary ß2-microglobulin secretion, all of which were significantly attenuated by PRO20. Urinary and renal levels of renin, angiotensinogen, and ANG II were elevated by AO and suppressed by PRO20, contrasting to largely unaltered plasma levels of the RAS parameters. The AO model also showed increased renal expression of full-length PRR and soluble PRR (sPRR) and urinary excretion of sPRR. Taken together, we conclude that PRR antagonism with PRO20 alleviates AO-induced nephropathy via inhibition of intrarenal RAS.


Subject(s)
Kidney Diseases/metabolism , Kidney/metabolism , Receptors, Cell Surface/metabolism , Renin-Angiotensin System , Serum Albumin, Bovine , Animals , Disease Models, Animal , Fibrosis , Inflammation Mediators/metabolism , Kidney/drug effects , Kidney/physiopathology , Kidney Diseases/chemically induced , Kidney Diseases/physiopathology , Kidney Diseases/prevention & control , Male , Nephrectomy , Oxidative Stress , Peptide Fragments/pharmacology , Proteinuria/chemically induced , Proteinuria/metabolism , Proteinuria/physiopathology , Proteinuria/prevention & control , Rats, Wistar , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics , Renin/pharmacology , Renin-Angiotensin System/drug effects , Signal Transduction/drug effects , Vacuolar Proton-Translocating ATPases , Prorenin Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...