Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Oncol Rep ; 48(1)2022 Jul.
Article in English | MEDLINE | ID: mdl-35642683

ABSTRACT

Following the publication of this paper, it was drawn to the Editors' attention by a concerned reader that the cell migration and invasion assay data shown in Figs. 2F, 5D and 6D were strikingly similar to data appearing in different form in other articles by different authors. Owing to the fact that the contentious data in the above article had already been published elsewhere, or were already under consideration for publication, prior to its submission to Oncology Reports, the Editor has decided that this paper should be retracted from the Journal. After having been in contact with the authors, they agreed with the decision to retract the paper. The Editor apologizes to the readership for any inconvenience caused. [the original article was published in Oncology Reports 38: 1857­1866, 2017; DOI: 10.3892/or.2017.5835].

2.
J Am Soc Nephrol ; 30(8): 1413-1424, 2019 08.
Article in English | MEDLINE | ID: mdl-31278195

ABSTRACT

BACKGROUND: Oxygen deprivation or hypoxia in the kidney drives CKD and contributes to end organ damage. The erythrocyte's role in delivery of oxygen (O2) is regulated by hypoxia, but the effects of CKD are unknown. METHODS: We screened all of the metabolites in the whole blood of mice infused with angiotensin II (Ang II) at 140 ng/kg per minute up to 14 days to simulate CKD and compared their metabolites with those from untreated mice. Mice lacking a receptor on their erythrocytes called ADORA2B, which increases O2 delivery, and patients with CKD were studied to assess the role of ADORA2B-mediated O2 delivery in CKD. RESULTS: Untargeted metabolomics showed increased production of 2,3-biphosphoglycerate (2,3-BPG), an erythrocyte-specific metabolite promoting O2 delivery, in mice given Ang II to induce CKD. Genetic studies in mice revealed that erythrocyte ADORA2B signaling leads to AMPK-stimulated activation of BPG mutase, promoting 2,3-BPG production and O2 delivery to counteract kidney hypoxia, tissue damage, and disease progression in Ang II-induced CKD. Enhancing AMPK activation in mice offset kidney hypoxia by triggering 2,3-BPG production and O2 delivery. Patients with CKD had higher 2,3-BPG levels, AMPK activity, and O2 delivery in their erythrocytes compared with controls. Changes were proportional to disease severity, suggesting a protective effect. CONCLUSIONS: Mouse and human evidence reveals that ADORA2B-AMPK signaling cascade-induced 2,3-BPG production promotes O2 delivery by erythrocytes to counteract kidney hypoxia and progression of CKD. These findings pave a way to novel therapeutic avenues in CKD targeting this pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Erythrocytes/metabolism , Hypoxia/metabolism , Kidney Failure, Chronic/metabolism , Oxygen/metabolism , Receptor, Adenosine A2B/metabolism , 2,3-Diphosphoglycerate/pharmacology , Adult , Angiotensin II/metabolism , Animals , Disease Progression , Female , Humans , Male , Metabolomics , Mice , Middle Aged , Models, Genetic , Signal Transduction
3.
Am J Hypertens ; 32(5): 476-485, 2019 04 22.
Article in English | MEDLINE | ID: mdl-30715101

ABSTRACT

BACKGROUND: Although numerous recent studies have shown a strong link between inflammation and hypertension, the underlying mechanisms by which inflammatory cytokines induce hypertension remain to be fully elucidated. Hypertensive disorders are also associated with elevated pressor sensitivity. Tissue transglutaminase (TG2), a potent cross-linking enzyme, is known to be transcriptionally activated by inflammatory cytokines and stabilize angiotensin II (Ang II) receptor AT1 (AT1R) via ubiquitination-preventing posttranslational modification. Here we sought to investigate the TG2-mediated AT1R stabilization in inflammation-induced hypertension and its functional consequences with a focus on receptor abundance and Ang II responsiveness. METHODS AND RESULTS: Using an experimental model of inflammation-induced hypertension established by introducing the pro-inflammatory tumor necrosis factor cytokine LIGHT, we provide pharmacologic and genetic evidence that TG2 is required for LIGHT-induced hypertension (systolic pressure on day 6: LIGHT = 152.3 ± 7.4 vs. LIGHT+ERW1041E [TG2 inhibitor] = 105.8 ± 13.1 or LIGHT+TG2-/- = 114.3 ± 4.3 mm Hg, P < 0.05, n = 4-5) and renal compromise (urine albumin/creatinine: LIGHT = 0.17 ± 0.05 vs. LIGHT+ERW1041E = 0.03 ± 0.01 or LIGHT+TG2-/- = 0.06 ± 0.01 mg/mg; plasma creatinine: LIGHT = 1.11 ± 0.04 vs. LIGHT+ERW1041E = 0.94 ± 0.04 or LIGHT+TG2-/- = 0.88 ± 0.09 mg/dl; urine volume: LIGHT = 0.23 ± 0.1 vs. LIGHT+ERW1041E = 0.84 ± 0.13 or LIGHT+TG2-/- = 1.02 ± 0.09 ml/24 hour on day 14, P < 0.05, n = 4-5). Our mechanistic studies showed that the TG2-mediated AT1R modification and accumulation (relative renal AT1R level: phosphate-buffered saline [PBS] = 1.23 ± 0.22, LIGHT = 3.49 ± 0.37, and LIGHT+ERW1041E = 1.77 ± 0.46, P < 0.05, n = 3; LIGHT+TG2+/+ = 85.28 ± 36.11 vs. LIGHT+TG2-/- = 7.01 ± 5.68, P < 0.05, n = 3) induced by LIGHT is associated with abrogated ß-arrestin binding (AT1R/associated ß-arrestin ratio: PBS = 2.62 ± 1.07, LIGHT = 38.60 ± 13.91, and LIGHT+ERW1041E = 6.97 ± 2.91, P < 0.05, n = 3; LIGHT+TG2+/+ = 66.43 ± 44.81 vs. LIGHT+TG2-/- = 2.45 ± 1.78, P < 0.01, n = 3) and could be found in renal medulla tubules of kidneys (relative tubular AT1R level: PBS = 5.91 ± 2.93, LIGHT = 92.82 ± 19.54, LIGHT+ERW1041E = 28.49 ± 11.65, and LIGHT+TG2-/- = 0.14 ± 0.10, P < 0.01, n = 5) and the blood vasculature (relative vascular AT1R level: PBS = 0.70 ± 0.30, LIGHT = 13.75 ± 2.49, and LIGHT+ERW1041E = 3.28 ± 0.87, P < 0.01, n = 3), 2 of the tissues highly related to the genesis of hypertension. Our in vitro cellular assays showed that LIGHT stimulation triggered a rapid TG2-dependent increase in the abundance of AT1Rs (relative AT1R level after 2-hour LIGHT treatment: AT1R (WT)+TG2 = 2.21 ± 0.23, AT1R (Q315A)+TG2 = 0.18 ± 0.23, P < 0.05 vs. starting point = 1, n = 2) and downstream calcium signaling (fold increase in NFAT-driven luciferase activity: Saline = 0.02 ± 0.03, Ang II = 0.17 ± 0.08, LIGHT = 0.05 ± 0.04, LIGHT+Ang II = 0.90 ± 0.04 (P < 0.01 vs. Ang II), and LIGHT+Ang II+ERW1041E = 0.15 ± 0.15 (P < 0.01 vs. LIGHT+Ang II), n = 3). CONCLUSIONS: Our data indicate an essential and systemic role for TG2 in bridging inflammation to hypertension via its posttranslational modifications stabilizing AT1 receptor and sensitizing Ang II. Our findings also suggest that TG2 inhibitors could be used as a novel group of cardiovascular agents.


Subject(s)
Blood Pressure/physiology , GTP-Binding Proteins/metabolism , Hypertension/metabolism , Inflammation/metabolism , Transglutaminases/metabolism , Animals , Disease Models, Animal , Female , Hypertension/etiology , Hypertension/physiopathology , Inflammation/complications , Kidney/metabolism , Male , Mice , Mice, Transgenic , Protein Glutamine gamma Glutamyltransferase 2 , Receptor, Angiotensin, Type 1/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/toxicity
4.
Oncol Rep ; 38(3): 1857-1866, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28731178

ABSTRACT

Lung cancer is one of the most common types of malignancy in humans and is a leading cause of cancer-related deaths among men and women worldwide. Aberrantly expressed microRNAs in non-small cell lung cancer (NSCLC) contribute to tumor occurrence and development as either tumor suppressors or promoters. MicroRNA-379 (miR­379) is dysregulated in several types of human cancer. However, its expression pattern, role and underlying mechanism in NSCLC progression and metastasis are poorly understood. In this study, assay of reverse transcription-quantitative polymerase chain reaction showed that miR­379 was downregulated in both NSCLC tissue and cell lines. Low miR­379 expression in NSCLC tissues was significantly correlated with TNM stage and lymph node metastasis. In addition, functional experiments revealed that restoring the expression of miR­379 inhibited cell proliferation, migration and invasion of NSCLC. The insulin-like growth factor receptor-1 (IGF­1R) was identified as a direct target of miR­379 in NSCLC. IGF­1R was highly expressed in NSCLC tissues and inversely correlated with miR­379 expression. Downregulation of IGF­1R had tumor suppressive roles similar to that of miR­379 overexpression on NSCLC cell proliferation, migration and invasion. Moreover, the upregulation of IGF­1R effectively rescued the tumor suppressive roles induced by miR­379 overexpression in NSCLC. The resumption of the expression of miR­379 inhibited the activation of AKT and ERK signaling pathways in NSCLC. These findings suggested that miR­379 acts as a tumor suppressor in NSCLC by directly targeting IGF­1R and indirectly regulating AKT and ERK signaling pathways. miR­379 provides novel therapeutic targets for the treatment of patients with this disease.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Genes, Tumor Suppressor/physiology , Lung Neoplasms/genetics , MAP Kinase Signaling System/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins c-akt/genetics , Receptors, Somatomedin/genetics , A549 Cells , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/pathology , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Male , Middle Aged , Receptor, IGF Type 1 , Signal Transduction/genetics
5.
Sci Rep ; 6: 29637, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27436223

ABSTRACT

Although Lands' cycle was discovered in 1958, its function and cellular regulation in membrane homeostasis under physiological and pathological conditions remain largely unknown. Nonbiased high throughput metabolomic profiling revealed that Lands' cycle was impaired leading to significantly elevated erythrocyte membrane lysophosphatidylcholine (LysoPC) content and circulating and erythrocyte arachidonic acid (AA) in mice with sickle cell disease (SCD), a prevalent hemolytic genetic disorder. Correcting imbalanced Lands' cycle by knockdown of phospholipase 2 (cPLA2) or overexpression of lysophosphatidycholine acyltransferase 1 (LPCAT1), two key enzymes of Lands' cycle in hematopoietic stem cells, reduced elevated erythrocyte membrane LysoPC content and circulating AA levels and attenuated sickling, inflammation and tissue damage in SCD chimeras. Human translational studies validated SCD mouse findings and further demonstrated that imbalanced Lands' cycle induced LysoPC production directly promotes sickling in cultured mouse and human SCD erythrocytes. Mechanistically, we revealed that hypoxia-mediated ERK activation underlies imbalanced Lands' cycle by preferentially inducing the activity of PLA2 but not LPCAT in human and mouse SCD erythrocytes. Overall, our studies have identified a pathological role of imbalanced Lands' cycle in SCD erythrocytes, novel molecular basis regulating Lands' cycle and therapeutic opportunities for the disease.


Subject(s)
Anemia, Sickle Cell/metabolism , Arachidonic Acid/blood , Erythrocytes/metabolism , Lysophosphatidylcholines/metabolism , Metabolomics/methods , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/genetics , Animals , Cell Hypoxia , Cells, Cultured , Disease Models, Animal , Female , Gene Knockdown Techniques , Group IV Phospholipases A2/genetics , Humans , Male , Mice
6.
J Am Heart Assoc ; 5(7)2016 06 30.
Article in English | MEDLINE | ID: mdl-27364991

ABSTRACT

BACKGROUND: The pathogenesis of essential hypertension is multifactorial with different underlying mechanisms contributing to disease. We have recently shown that TNF superfamily member 14 LIGHT (an acronym for homologous to lymphotoxins, exhibits inducible expression, and competes with herpes simplex virus glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes, also known as TNFSF14) induces hypertension when injected into mice. Research reported here was undertaken to examine the role of transglutaminase (TGase) in LIGHT-induced hypertension. METHODS AND RESULTS: Initial experiments showed that plasma and kidney TGase activity was induced by LIGHT infusion (13.91±2.92 versus 6.75±1.92 mU/mL and 19.86±3.55 versus 12.00±0.97 mU/10 µg) and was accompanied with hypertension (169±7.16 versus 117.17±11.57 mm Hg at day 14) and renal impairment (proteinuria, 61.33±23.21 versus 20.38±9.01 µg/mg; osmolality, 879.57±93.02 versus 1407.2±308.04 mmol/kg). The increase in renal TGase activity corresponded to an increase in RNA for the tissue TGase isoform, termed TG2. Pharmacologically, we showed that LIGHT-induced hypertension and renal impairment did not occur in the presence of cystamine, a well-known competitive inhibitor of TGase activity. Genetically, we showed that LIGHT-mediated induction of TGase, along with hypertension and renal impairment, was dependent on interleukin-6 and endothelial hypoxia inducible factor-1α. We also demonstrated that interleukin-6, endothelial hypoxia inducible factor-1α, and TGase are required for LIGHT-induced production of angiotensin receptor agonistic autoantibodies. CONCLUSIONS: Thus, LIGHT-induced hypertension, renal impairment, and production of angiotensin receptor agonistic autoantibodies require TGase, most likely the TG2 isoform. Our findings establish TGase as a critical link between inflammation, hypertension, and autoimmunity.


Subject(s)
Blood Pressure/drug effects , Hypertension/immunology , Inflammation/immunology , Transglutaminases/drug effects , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology , Animals , Autoantibodies/immunology , Blood Pressure/immunology , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Hypertension/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/metabolism , Interleukin-6/genetics , Interleukin-6/immunology , Kidney/immunology , Kidney/metabolism , Mice , Mice, Knockout , Protein Glutamine gamma Glutamyltransferase 2 , Proteinuria/immunology , Proteinuria/metabolism , Receptors, Angiotensin/immunology , Renal Insufficiency/immunology , Renal Insufficiency/metabolism , Transglutaminases/genetics , Transglutaminases/metabolism
7.
J Am Heart Assoc ; 4(12)2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26675250

ABSTRACT

BACKGROUND: Preeclampsia (PE) is a life-threatening hypertensive disorder of pregnancy associated with autoantibodies, termed AT1-AA, that activate the AT1 angiotensin receptor. Although the pathogenic nature of these autoantibodies has been extensively studied, little is known about the molecular cause of their generation. METHODS AND RESULTS: Here we show that tissue transglutaminase (TG2), an enzyme that conducts posttranslational modification of target proteins, directly modified the 7-amino acid (7-aa) epitope peptide that localizes to the second extracellular loop of the AT1 receptor. These findings led us to further discover that plasma transglutaminase activity was induced and contributed to the production of AT1-AA and disease development in an experimental model of PE induced by injection of LIGHT, a tumor necrosis factor superfamily member. Key features of PE were regenerated by adoptive transfer of purified IgG from LIGHT-injected pregnant mice and blocked by the 7-amino acid epitope peptide. Translating our mouse research to humans, we found that plasma transglutaminase activity was significantly elevated in PE patients and was positively correlated with AT1-AA levels and PE features. CONCLUSIONS: Overall, we provide compelling mouse and human evidence that elevated transglutaminase underlies AT1-AA production in PE and highlight novel pathogenic biomarkers and innovative therapeutic possibilities for the disease.


Subject(s)
Autoantibodies/immunology , GTP-Binding Proteins/physiology , Pre-Eclampsia/etiology , Receptors, Angiotensin/physiology , Transglutaminases/physiology , Adult , Animals , Biomarkers/blood , Blotting, Western , Epitopes/immunology , Female , GTP-Binding Proteins/blood , Humans , Mice , Mice, Inbred C57BL , Pre-Eclampsia/physiopathology , Pregnancy , Protein Glutamine gamma Glutamyltransferase 2 , Receptor, Angiotensin, Type 1/physiology , Transglutaminases/blood
8.
Hypertension ; 66(1): 75-84, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25987665

ABSTRACT

Hypertensive chronic kidney disease is one of the most prevalent medical conditions with high morbidity and mortality in the United States and worldwide. However, early events initiating the progression to hypertensive chronic kidney disease are poorly understood. We hypothesized that elevated endothelial hypoxia-inducible factor-1α (HIF-1α) is a common early insult triggering initial glomerular injury leading to hypertensive chronic kidney disease. To test our hypothesis, we used an angiotensin II infusion model of hypertensive chronic kidney disease to determine the specific cell type and mechanisms responsible for elevation of HIF-1α and its role in the progression of hypertensive chronic kidney disease. Genetic studies coupled with reverse transcription polymerase chain reaction profiling revealed that elevated endothelial HIF-1α is essential to initiate glomerular injury and progression to renal fibrosis by the transcriptional activation of genes encoding multiple vasoactive proteins. Mechanistically, we found that endothelial HIF-1α gene expression was induced by angiotensin II in a nuclear factor-κB-dependent manner. Finally, we discovered reciprocal positive transcriptional regulation of endothelial Hif-1α and Nf-κb genes is a key driving force for their persistent activation and disease progression. Overall, our findings revealed that the stimulation of HIF-1α gene expression in endothelial cells is detrimental to induce kidney injury, hypertension, and disease progression. Our findings highlight early diagnostic opportunities and therapeutic approaches for hypertensive chronic kidney disease.


Subject(s)
Hypertension/complications , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Kidney Glomerulus/pathology , Renal Insufficiency, Chronic/etiology , Angiotensin II/pharmacology , Angiotensin II/toxicity , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Cadherins/biosynthesis , Cadherins/genetics , Cells, Cultured , Disease Progression , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelin-1/blood , Endothelium, Vascular/metabolism , Feedback, Physiological , Fibrosis , Gene Expression Regulation/drug effects , Humans , Hypertension/chemically induced , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney Glomerulus/metabolism , Mice , Mice, Transgenic , NF-kappa B/biosynthesis , NF-kappa B/genetics , NF-kappa B/metabolism , Organ Culture Techniques , RNA, Small Interfering/pharmacology , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/physiopathology , Up-Regulation/drug effects
9.
Biochem Biophys Res Commun ; 461(1): 186-92, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25881503

ABSTRACT

Renal interstitial fibrosis closely relates to chronic kidney disease and is regarded as the final common pathway in most cases of end-stage renal disease. Metabolomic biomarkers can facilitate early diagnosis and allow better understanding of the pathogenesis underlying renal fibrosis. Gas chromatography-mass spectrometry (GC/MS) is one of the most promising techniques for identification of metabolites. However, the existence of the background, baseline offset, and overlapping peaks makes accurate identification of the metabolites unachievable. In this study, GC/MS coupled with chemometric methods was successfully developed to accurately identify and seek metabolic biomarkers for rats with renal fibrosis. By using these methods, seventy-six metabolites from rat serum were accurately identified and five metabolites (i.e., urea, ornithine, citric acid, galactose, and cholesterol) may be useful as potential biomarkers for renal fibrosis.


Subject(s)
Algorithms , Biomarkers/blood , Blood Chemical Analysis/methods , Data Interpretation, Statistical , Gas Chromatography-Mass Spectrometry/methods , Kidney/metabolism , Renal Insufficiency, Chronic/blood , Animals , Fibrosis/blood , Male , Multivariate Analysis , Rats , Rats, Wistar , Renal Insufficiency, Chronic/diagnosis , Reproducibility of Results , Sensitivity and Specificity
10.
Tumour Biol ; 35(12): 11727-33, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25374059

ABSTRACT

To date, several studies have been conducted to assess the association between endothelial nitric oxide synthase (eNOS) gene 894G > T polymorphism and prostate cancer (PCa) risk, but the results are conflicting. To derive a more precise estimation of the relationship between 894G > T polymorphism and PCa risk, the present meta-analysis was performed. A total of eight case-control studies were included in this meta-analysis. The pooled odds ratio (OR) with 95 % confidence interval (CI) was calculated to evaluate the associations. Our results suggested that 894G > T polymorphism is associated with PCa risk under codominant (GT vs. GG) (OR = 1.11, 95 % CI = 1.01-1.22, P = 0.04) and overdominant (GT vs. GG + TT) (OR = 1.12, 95 % CI = 1.02-1.23, P = 0.02) models in the overall population, while there are no associations observed under dominant (GT + TT vs. GG), recessive (TT vs. GG + GT), and allelic (T vs. G) models. Moreover, when the eligible studies were stratified according to sources of control, significant association between 894G > T polymorphism and susceptibility of PCa was also identified under codominant (OR = 1.12, 95 % CI = 1.01-1.24, P = 0.03) and overdominant (OR = 1.13, 95 % CI = 1.02-1.25, P = 0.02) models when using healthy individuals as control. However, there are no significant associations found under any genetic models when using BPH patients as control group. In conclusion, the present meta-analysis suggested that the eNOS gene 894G > T polymorphism might be a risk factor in the onset of PCa.


Subject(s)
Genetic Predisposition to Disease , Nitric Oxide Synthase Type III/genetics , Polymorphism, Single Nucleotide , Prostatic Neoplasms/genetics , Alleles , Case-Control Studies , Gene Frequency , Genetic Association Studies , Genotype , Humans , Male , Odds Ratio , Publication Bias
11.
PLoS One ; 9(10): e111242, 2014.
Article in English | MEDLINE | ID: mdl-25347392

ABSTRACT

Diabetic nephropathy (DN) remains the leading cause of end-stage renal disease (ESRD), a situation that is in part attributable to the lack of effective treatments. Fluorofenidone is a newly developed reagent with anti-fibrotic activity. While fluorofenidone was previously demonstrated to possess renoprotection from DN pathogenesis in db/db mice, the protective process and its underlying mechanisms have not been well studied. To characterize fluorofenidone-derived renoprotection, we treated 5, 8, or 12-week old db/db mice with daily doses of placebo, fluorofenidone, or losartan until 24 weeks of age; the time at which diabetes and DN were fully developed in placebo-treated animals. In comparison to db/db mice receiving fluorofenidone at 12-weeks old, those treated at 5-weeks had less glomerular expansion and better preservation of renal functions, judged by serum creatinine levels, albumin to creatinine ratio, and urinary albumin excretion (mg/24 hours). These benefits of early treatment were associated with significant reductions of multiple DN-promoting events, such as decreased expression of TGF-ß1 and the p22phox subunit of NADPH oxidase as well as downregulated activation of protein kinase C-zeta (ζ), ERK and AKT. This improvement in renoprotection following early interventions is not a unique property of DN pathogenesis, as losartan does not apparently offer the same benefits and is not more renoprotective than fluorofenidone. Additionally, the enhanced renoprotection provided by fluorofenidone did not affect the diabetic process, as it did not alter serum levels of glycated serum proteins, glucose, triglyceride or cholesterol. Collectively, we provide evidence that fluorofenidone offers improved renoprotection at early stages of DN pathogenesis.


Subject(s)
Diabetic Neuropathies/drug therapy , Kidney/drug effects , Pyridones/therapeutic use , Animals , Blood Glucose/metabolism , Blood Proteins/metabolism , Cholesterol/blood , Kidney/metabolism , Male , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , NADPH Oxidases/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyridones/pharmacology , Transforming Growth Factor beta/metabolism
12.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 39(6): 591-7, 2014 Jun.
Article in Chinese | MEDLINE | ID: mdl-25011972

ABSTRACT

OBJECTIVE: To determine the effect of curcumin on diabetic nephropathy in db/db mice and its possible mechanism. METHODS: Ten female db/db mice were randomly divided into 2 groups: one was treated with curcumin at 200 mg/(kg.d) and the other was a placebo group. Five age-matched db/m mice were grouped as the controls. In the curcumin group, curcumin was administered to db/db mice for 18 weeks. At the end of the experiment, the blood glucose and albumin were measured, and the kidney tissue sections were stained with PAS to observe the pathological changes. The expression of collagen IV and FN in the kidney was detected by immunohitochemistry staining. Western blot was used to detect the phosphorylation of signal transducer and activator of transcription 3 (STAT3) and IκB in the kidney. RESULTS: Compared with db/m mice, the weight and blood glucose of db/db mice were markedly increased, accompanied with heavy proteinuria, glomerulus hypertrophy, mesangial area expansion, thickening of basement membrane and ECM deposition. The phosphorylation of STAT3 was upregulated and the degradation of IκB was increased. Compared with the db/db mice, curcumin significantly decreased the urinary albumin, inhibited the phosphorylation of STAT3 and the degradation of IκB, and reduced the expression of collagen IV and FN in the kidney. CONCLUSION: Curcumin can obviously decrease albuminuria and attenuate glomerular sclerosis in diabetic db/db mice by inhibiting phosphorylation of STAT3 and degradation of IκB.


Subject(s)
Curcumin/pharmacology , Diabetic Nephropathies/drug therapy , I-kappa B Proteins/metabolism , STAT3 Transcription Factor/metabolism , Albuminuria , Animals , Blood Glucose , Collagen Type IV/metabolism , Diabetes Mellitus, Type 2 , Female , Fibronectins/metabolism , Kidney/drug effects , Kidney/metabolism , Mice , Phosphorylation , Proteinuria
13.
Nephrology (Carlton) ; 19(8): 479-89, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24750539

ABSTRACT

AIM: Apoptosis is one of the most important mechanisms underlying renal interstitial fibrosis. We identified the role of protein Niban in apoptosis of tumour cells. The purpose of this study was to assess the expression of Niban in renal interstitial fibrosis of humans and rats. METHODS: Immunohistochemistry was used to detect Niban in patients with obstructive nephropathy. Proteomics and gene array analysis were performed to screen different molecules involved in the pathophysiology of unilateral-ureteral obstruction rats. We confirmed Niban using immunohistochemistry and Western blot in renal cortex of UUO rats and HK-2 cells. TUNEL assay and flow cytometry revealed apoptosis of renal tubular cells. siRNA and overexpression plasmid were transfected specifically to study the possible function of Niban. RESULTS: Niban was decreased apparently in renal tubular cells of patients with obstructive nephropathy, compared with controls. Niban decreased in renal cortex of UUO rats and transforming growth factor-ß1 (TGF-ß1)-stimulated HK-2 cells. siRNA of Niban increased apoptosis of HK-2 cells. TGF-ß1 also increased apoptosis of HK-2 cells. Overexpression of Niban failed to diminish apoptosis of HK-2 cells induced by TGF-ß1. CONCLUSIONS: Niban decreased in renal tubular cells of patients of obstructive nephropathy, UUO rats and TGF-ß1 stimulated HK-2 cells. Suppressing Niban increases apoptosis in HK-2 cells. Niban may be associated with apoptosis of HK-2 cells.


Subject(s)
Biomarkers, Tumor/biosynthesis , Kidney/metabolism , Kidney/pathology , Neoplasm Proteins/biosynthesis , Animals , Apoptosis , Biomarkers, Tumor/analysis , Cells, Cultured , Fibrosis/genetics , Fibrosis/metabolism , Humans , Kidney/chemistry , Male , Neoplasm Proteins/analysis , Rats , Rats, Sprague-Dawley
14.
Hypertension ; 63(3): 595-606, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24324043

ABSTRACT

Preeclampsia, a prevalent hypertensive disorder of pregnancy, is believed to be secondary to uteroplacental ischemia. Accumulating evidence indicates that hypoxia-independent mediators, including inflammatory cytokines and growth factors, are associated with preeclampsia, but it is unclear whether these signals directly contribute to placental damage and disease development in vivo. We report that LIGHT, a novel tumor necrosis factor superfamily member, is significantly elevated in the circulation and placentas of preeclamptic women compared with normotensive pregnant women. Injection of LIGHT into pregnant mice induced placental apoptosis, small fetuses, and key features of preeclampsia, hypertension and proteinuria. Mechanistically, using neutralizing antibodies specific for LIGHT receptors, we found that LIGHT receptors herpes virus entry mediator and lymphotoxin ß receptor are required for LIGHT-induced placental impairment, small fetuses, and preeclampsia features in pregnant mice. Accordingly, we further revealed that LIGHT functions through these 2 receptors to induce secretion of soluble fms-like tyrosine kinase-1 and endothelin-1, 2 well-accepted pathogenic factors in preeclampsia, and thereby plays an important role in hypertension and proteinuria in pregnant mice. Lastly, we extended our animal findings to human studies and demonstrated that activation of LIGHT receptors resulted in increased apoptosis and elevation of soluble fms-like tyrosine kinase-1 secretion in human placental villous explants. Overall, our human and mouse studies show that LIGHT signaling is a previously unrecognized pathway responsible for placental apoptosis, elevated secretion of vasoactive factors, and subsequent maternal features of preeclampsia, and reveal new therapeutic opportunities for the management of the disease.


Subject(s)
Endothelins/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , Pregnancy, Animal , Proteinuria/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Adult , Animals , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , Pre-Eclampsia/physiopathology , Pregnancy , Proteinuria/etiology , Signal Transduction
15.
Circ Res ; 112(11): 1466-78, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23584256

ABSTRACT

RATIONALE: Hypertension is the most prevalent life-threatening disease worldwide and is frequently associated with chronic kidney disease (CKD). However, the molecular basis underlying hypertensive CKD is not fully understood. OBJECTIVE: We sought to identify specific factors and signaling pathways that contribute to hypertensive CKD and thereby exacerbate disease progression. METHODS AND RESULTS: Using high-throughput quantitative reverse-transcription polymerase chain reaction profiling, we discovered that the expression level of 5'-ectonucleotidase (CD73), a key enzyme that produces extracellular adenosine, was significantly increased in the kidneys of angiotensin II-infused mice, an animal model of hypertensive nephropathy. Genetic and pharmacological studies in mice revealed that elevated CD73-mediated excess renal adenosine preferentially induced A2B adenosine receptor (ADORA2B) production and that enhanced kidney ADORA2B signaling contributes to angiotensin II-induced hypertension. Similarly, in humans, we found that CD73 and ADORA2B levels were significantly elevated in the kidneys of CKD patients compared with normal individuals and were further elevated in hypertensive CKD patients. These findings led us to further discover that elevated renal CD73 contributes to excess adenosine signaling via ADORA2B activation that directly stimulates endothelin-1 production in a hypoxia-inducible factor-α-dependent manner and underlies the pathogenesis of the disease. Finally, we revealed that hypoxia-inducible factor-α is an important factor responsible for angiotensin II-induced CD73 and ADORA2B expression at the transcriptional level. CONCLUSIONS: Overall, our studies reveal that angiotensin II-induced renal CD73 promotes the production of renal adenosine that is a prominent driver of hypertensive CKD by enhanced ADORA2B signaling-mediated endothelin-1 induction in a hypoxia-inducible factor-α-dependent manner. The inhibition of excess adenosine-mediated ADORA2B signaling represents a novel therapeutic target for the disease.


Subject(s)
5'-Nucleotidase/metabolism , Hypertension, Renal/metabolism , Kidney/metabolism , Receptor, Adenosine A2B/metabolism , 5'-Nucleotidase/genetics , Adenosine/metabolism , Adult , Angiotensin II/pharmacology , Animals , Cells, Cultured , Chronic Disease , Endothelial Cells/cytology , Endothelial Cells/physiology , Endothelin-1/metabolism , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression/physiology , Humans , Hypertension, Renal/chemically induced , Hypertension, Renal/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Receptor, Adenosine A2B/genetics , Signal Transduction/physiology , Vasoconstrictor Agents/pharmacology
16.
Molecules ; 17(1): 884-96, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22252504

ABSTRACT

Pirfenidone, a pyridone compound, is an effective and novel antifibrotic agent. In this article, we describe the design, synthesis and activity evaluation of novel antifibrotic agents, 1-(substituted aryl)-5-trifluoromethyl-2(1H) pyridones modified with carbohydrate. Most of the title compounds exhibited comparable or better inhibitory activity than fluorofenidone. Notably, compound 19a demonstrated the highest cell-based inhibitory activity against NIH 3T3 (IC(50) = 0.17 mM).


Subject(s)
Cell Proliferation/drug effects , Monosaccharides/chemistry , Pyridones/chemical synthesis , Pyridones/pharmacology , Animals , Drug Design , Fibrosis/drug therapy , Hydrophobic and Hydrophilic Interactions , Inhibitory Concentration 50 , Mice , NIH 3T3 Cells , Pyridones/chemistry , Solubility
17.
Zhonghua Xin Xue Guan Bing Za Zhi ; 39(8): 743-8, 2011 Aug.
Article in Chinese | MEDLINE | ID: mdl-22169423

ABSTRACT

OBJECTIVE: To investigate the effect of rosuvastatin on atherosclerosis in apoE-knockout (apoE-/-) mice. METHODS: Eighteen 6-week-old apoE-/- mice fed with high fat diet were used as atherosclerosis models, twelve 6-week-old C57BL/6 mice fed with normal diet were used as control. After twelve weeks, six apoE-/- mice were used to observe the formation of atherosclerosis. Another 12 apoE-/- mice were divided into placebo treated group (n = 6) and rosuvastatin group (n = 6, 10 mg×kg(-1)×d(-1) per gavage) and treated for 12 weeks. Then, blood was collected for measuring lipid, aorta was prepared for morphologic study (HE, Oil red O, Masson) and immunohistochemical analysis (α-smooth active protein, transforming growth factor ß(1), macrophage surface molecule-3). RESULTS: Serum cholesterol and low density lipoprotein levels were significantly higher in apoE-/- mice fed with high fat diet than in C57/BL6 mice(all P < 0.01)while triglyceride level was similar between the two groups, these were not affected by rosuvastatin. Similarly, atherosclerotic lesion area in apoE-/- mice fed with high fat diet was also not significantly reduced by rosuvastatin, while lipid deposition could be significantly reduced and collagen deposition could be significantly increased in the aortic atherosclerotic lesions by treatment with rosuvastatin. Upregulated TGF-ß(1) and Mac-3 expression in the aortic atherosclerotic lesions in apoE-/- mice fed with high fat diet could also be significantly reduced by rosuvastatin (all P < 0.01), suggesting reduce inflammatory responses in the atherosclerotic lesion and stable atherosclerotic plaque post rosuvastatin treatment. CONCLUSION: Reducing inflammatory responses and stabilizing plaque properties might contribute to the anti-atherosclerosis effects of rosuvastatin in mice high fat diet fed apoE-/- mice.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Fluorobenzenes/pharmacology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Antigens, Differentiation/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Diet, High-Fat , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/pathology , Rosuvastatin Calcium , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...