Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Int J Mol Sci ; 24(11)2023 Jun 04.
Article in English | MEDLINE | ID: mdl-37298689

ABSTRACT

Among kidney cancers, clear cell renal cell carcinoma (ccRCC) has the highest incidence rate in adults. The survival rate of patients diagnosed as having metastatic ccRCC drastically declines even with intensive treatment. We examined the efficacy of simvastatin, a lipid-lowering drug with reduced mevalonate synthesis, in ccRCC treatment. Simvastatin was found to reduce cell viability and increase autophagy induction and apoptosis. In addition, it reduced cell metastasis and lipid accumulation, the target proteins of which can be reversed through mevalonate supplementation. Moreover, simvastatin suppressed cholesterol synthesis and protein prenylation that is essential for RhoA activation. Simvastatin might also reduce cancer metastasis by suppressing the RhoA pathway. A gene set enrichment analysis (GSEA) of the human ccRCC GSE53757 data set revealed that the RhoA and lipogenesis pathways are activated. In simvastatin-treated ccRCC cells, although RhoA was upregulated, it was mainly restrained in the cytosolic fraction and concomitantly reduced Rho-associated protein kinase activity. RhoA upregulation might be a negative feedback effect owing to the loss of RhoA activity caused by simvastatin, which can be restored by mevalonate. RhoA inactivation by simvastatin was correlated with decreased cell metastasis in the transwell assay, which was mimicked in dominantly negative RhoA-overexpressing cells. Thus, owing to the increased RhoA activation and cell metastasis in the human ccRCC dataset analysis, simvastatin-mediated Rho inactivation might serve as a therapeutic target for ccRCC patients. Altogether, simvastatin suppressed the cell viability and metastasis of ccRCC cells; thus, it is a potentially effective ccRCC adjunct therapy after clinical validation for ccRCC treatment.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Simvastatin/pharmacology , Carcinoma, Renal Cell/drug therapy , Mevalonic Acid/metabolism , Kidney Neoplasms/drug therapy , Lipids , rhoA GTP-Binding Protein/metabolism
2.
Front Med (Lausanne) ; 9: 997379, 2022.
Article in English | MEDLINE | ID: mdl-36203771

ABSTRACT

Whether valacyclovir-associated neurotoxicity (VAN) occurs more frequently in patients with end-stage renal disease (ESRD) on dialysis is unknown. This is the first population-based study to examine the risk of VAN associated with ESRD patients on dialysis. Among 2,284,800 patients diagnosed as having herpes zoster from 2002 to 2016, patients with ESRD on dialysis and individuals with normal renal function were enrolled in this study. Following propensity score matching, we compared the risk of altered mental status between valacyclovir users and non-users in the ESRD and normal renal function cohorts over a 30-day follow-up period. In the ESRD cohort, the incidence of altered mental status was 1.68 and 0.52 per 1,000 person-day in valacyclovir users and non-users, respectively, with an adjusted hazard ratio (HR) of 3.22 (95% confidence interval [CI]: 2.04-4.99, P < 0.001). The incidence of altered mental status of valacyclovir users on hemodialysis (HD) and peritoneal dialysis (PD) was higher than that of non-users. The adjusted HR was 3.20 (95% CI: 1.98-5.15, P < 0.001) for those on HD and 3.44 (95% CI: 1.13-10.49, P = 0.030) for those with PD. However, altered mental status was not observed in patients on HD receiving ≤500 mg of valacyclovir three times per week or in those on PD receiving ≤500 mg of valacyclovir per day. The findings demonstrate that adjusting the valacyclovir dosage and monitoring VAN in patients with HD and PD who have herpes zoster is crucial.

3.
Article in English | MEDLINE | ID: mdl-35966750

ABSTRACT

Diabetes in children and its complications are on the rise globally, which is accompanied by increasing in diabetes-related complications. Oxidative stress and inflammation induced by elevated blood sugar in diabetic patients are considered risk factors associated with the development of diabetes complications, including chronic kidney disease and its later development to end-stage renal disease. Microvascular changes within the kidneys of DM patients often lead to chronic kidney disease, which aggravates the illness. Sigesbeckia orientalis extract (SOE), reported to have strong antioxidative and excellent anti-inflammatory activities, is used in the modern practice of traditional Chinese medicine. Kidneys from three groups of control mice (CTR), mice with streptozotocin (STZ)-induced diabetes (DM), and mice with STZ-induced DM treated with SOE (DMRx) were excised for morphological analyses and immunohistochemical assessments. Only mice in the DM group exhibited significantly lower body weight, but higher blood sugar was present. The results revealed more obvious renal injury in the DM group than in the other groups, which appeared as greater glomerular damage and tubular injury, sores, and plenty of connective tissues within the mesangium. Not only did the DM group have a higher level of cytokine, tumor necrosis factor, and the oxidative stress marker, 8-hydroxyguanosine expression, but also factors of the nuclear factor pathway and biomarkers of microvascular status had changed. Disturbances to the kidneys in DMRx mice were attenuated compared to the DM group. We concluded that SOE is an effective medicine, with antioxidative and anti-inflammatory abilities, to protect against or attenuate diabetic nephropathy from inflammatory disturbances by oxidative stress and to cure vessel damage in a hyperglycemic situation.

4.
Front Oncol ; 12: 840142, 2022.
Article in English | MEDLINE | ID: mdl-35600378

ABSTRACT

Objective: Plasma dipeptidyl peptidase-4 (DPP4) levels were significantly lower in patients with colorectal and liver cancers, and animal studies also showed DPP4 inhibitors (DPP4is) have procarcinogenic effects in colorectal cancer. Until now, whether DPP4is therapy affects the progression of liver cancer and colorectal cancer in patients with T2DM has not been well investigated. We investigated the association between cumulative defined daily dose (cDDD) of DPP4is exposure and risks of liver and colorectal cancers in patients with type 2 diabetes. Materials and Methods: We identified 268,520 patients with diabetes receiving DPP4is as second-line agents between March 1, 2009, and December 31, 2013, from Taiwan's National Health Insurance Research Database, Taiwan Cancer Registry, and National Death Registry of Taiwan. The amount of DPP4is were divided into three groups (low, medium, and high) based on the interquartile range of the cDDD of the DPP4is. Results: The data showed that the low cDDD of DPP-4is was associated with a reducing risk of colorectal cancer [adjusted odds ratio (OR), 0.49; 95% CI, 0.32-0.75; P=0.001]. However, the high cDDD of DPP-4is was associated with an increasing risk of colorectal cancer (adjusted OR, 1.86; 95% CI, 1.32-2.61; P<0.001). No association between DPP4is use and liver cancer risk was observed. Conclusions: This nested case study revealed a J-shaped association between the cDDD of DPP-4is and colorectal cancer risk, but not liver cancer risk. Therefore, the effects of long-term DPP4is use on colorectal cancer risk warrant further study.

5.
Sci Rep ; 12(1): 4673, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35304586

ABSTRACT

We previously reported that perfluorooctanesulfonate (PFOS) causes autophagy-induced apoptosis in renal tubular cells (RTCs) through a mechanism dependent on reactive oxygen species (ROS)/extracellular signal-regulated kinase. This study extended our findings and determined the therapeutic potency of L-Carnitine in PFOS-treated RTCs. L-Carnitine (10 mM) reversed the effects of PFOS (100 µM) on autophagy induction and impaired autophagy flux. Furthermore, it downregulated the protein level of p47Phox, which is partly related to PFOS-induced increased cytosolic ROS in RTCs. Moreover, L-Carnitine reduced ROS production in mitochondria and restored PFOS-impeded mitochondrial function, leading to sustained normal adenosine triphosphate synthesis and oxygen consumption and reduced proton leakage in a Seahorse XF stress test. The increased inositol-requiring enzyme 1α expression by PFOS, which indicated endoplasmic reticulum (ER) stress activation, was associated with PFOS-mediated autophagy activation that could be attenuated through 4-phenylbutyrate (5 mM, an ER stress inhibitor) and L-Carnitine pretreatment. Therefore, by reducing the level of IRE1α, L-Carnitine reduced the levels of Beclin and LC3BII, consequently reducing the level of apoptotic biomarkers including Bax and cleaving PARP and caspase 3. Collectively, these results indicate that through the elimination of oxidative stress, extracellular signal-regulated kinase activation, and ER stress, L-Carnitine reduced cell autophagy/apoptosis and concomitantly increased cell viability in RTCs. This study clarified the potential mechanism of PFOS-mediated RTC apoptosis and provided a new strategy for using L-Carnitine to prevent and treat PFOS-induced RTC apoptosis.


Subject(s)
Endoplasmic Reticulum Stress , Endoribonucleases , Alkanesulfonic Acids , Apoptosis , Autophagy , Carnitine/pharmacology , Extracellular Signal-Regulated MAP Kinases , Fluorocarbons , Mitochondria/metabolism , Protein Serine-Threonine Kinases , Reactive Oxygen Species/metabolism
6.
Int J Mol Sci ; 22(23)2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34884829

ABSTRACT

Methylprednisolone (MP) is an anti-inflammatory drug approved for the treatment of acute spinal cord injuries (SCIs). However, MP administration for SCIs has become a controversial issue while the molecular effects of MP remain unexplored to date. Therefore, delineating the benefits and side effects of MP and determining what MP cannot cure in SCIs at the molecular level are urgent issues. Here, genomic profiles of the spinal cord in rats with and without injury insults, and those with and without MP treatment, were generated at 0, 2, 4, 6, 8, 12, 24, and 48 h post-injury. A comprehensive analysis was applied to obtain three distinct classes: side effect of MP (SEMP), competence of MP (CPMP), and incapability of MP (ICMP). Functional analysis using these genes suggested that MP exerts its greatest effect at 8~12 h, and the CPMP was reflected in the immune response, while SEMP suggested aspects of metabolism, such as glycolysis, and ICMP was on neurological system processes in acute SCIs. For the first time, we are able to precisely reveal responsive functions of MP in SCIs at the molecular level and provide useful solutions to avoid complications of MP in SCIs before better therapeutic drugs are available.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Methylprednisolone/pharmacology , Spinal Cord Injuries/pathology , Transcriptome/drug effects , Animals , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal , Female , Methylprednisolone/therapeutic use , Rats , Rats, Long-Evans , Spinal Cord/metabolism , Spinal Cord Injuries/drug therapy , Time Factors
7.
PLoS One ; 16(1): e0245442, 2021.
Article in English | MEDLINE | ID: mdl-33471797

ABSTRACT

Perfluorooctane sulfonate (PFOS) is among the most abundant organic pollutants and is widely distributed in the environment, wildlife, and humans. Its toxic effects and biological hazards are associated with its long elimination half-life in humans. However, how it affects renal tubular cells (RTCs) remains unclear. In this study, PFOS was observed to mediate the increase in reactive oxygen species (ROS) generation, followed by the activation of the extracellular-signal-regulated kinase 1/2 (ERK1/2) pathway, which induced autophagy in RTCs. Although PFOS treatment induced autophagy after 6 h, prolonged treatment (24 h) reduced the autophagic flux by increasing lysosomal membrane permeability (LMP), leading to increased p62 protein accumulation and subsequent apoptosis. The increase in LMP was visualized through increased green fluorescence with acridine orange staining, and this was attenuated by 3-methyladenine, an autophagy inhibitor. N-acetyl cysteine and an inhibitor of the mitogen-activated protein kinase kinases (U0126) attenuated autophagy and apoptosis. Taken together, these results indicate that ROS activation and ROS-mediated phosphorylated ERK1/2 activation are essential to activate autophagy, resulting in the apoptosis of PFOS-treated RTCs. Our findings provide insight into the mechanism of PFOS-mediated renal toxicity.


Subject(s)
Alkanesulfonic Acids/toxicity , Apoptosis/drug effects , Environmental Pollutants/toxicity , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorocarbons/toxicity , Kidney Tubules/drug effects , Oxidative Stress/drug effects , Animals , Autophagy/drug effects , Cell Line , Enzyme Activation/drug effects , Kidney Tubules/cytology , Kidney Tubules/metabolism , Rats
8.
Nutrients ; 12(9)2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32899821

ABSTRACT

The effects of ketoanalogues (KA) supplementation on mortality and progression to dialysis in patients with pre-dialysis stage 5 chronic kidney disease (CKD) receiving a low-protein diet (LPD) remain ambiguous. From Taiwan's National Health Insurance Research Database during 1996-2011, 165 patients with pre-dialysis CKD on an LPD (0.6 g/kg/day) with KA supplementation were matched with 165 patients with pre-dialysis CKD on an LPD without KA supplementation. Of the 165 patients with advanced CKD receiving KA supplementation, 34 (20.6%) died, and 124 (75.2%) underwent long-term dialysis during the study period. There was no significant difference in mortality between the KA-user group and the KA-nonuser group (adjusted hazard ratio [HR], 1.41; 95% confidence interval [CI], 0.68-2.93; p = 0.355). KA supplementation significantly increased long-term dialysis risk (adjusted HR, 1.41; 95% CI, 1.04-1.90; p = 0.025) and combined outcome risk (defined as long-term dialysis and death; adjusted HR, 1.37; 95% CI, 1.02-1.83; p = 0.034). KA supplementation also increased long-term dialysis risk (adjusted HR, 1.49; 95% CI, 1.00-2.20; p = 0.048) in the subgroup of pre-dialysis patients with diabetes mellitus (DM), but not in those patients without DM. In conclusion, KA supplementation might increase long-term dialysis risk in patients with advanced CKD receiving an LPD, but it did not increase mortality.


Subject(s)
Diet, Protein-Restricted/mortality , Dietary Supplements , Keto Acids/administration & dosage , Renal Dialysis/mortality , Renal Insufficiency, Chronic/mortality , Databases, Factual , Disease Progression , Female , Humans , Kidney/physiopathology , Male , Middle Aged , Proportional Hazards Models , Renal Insufficiency, Chronic/therapy , Taiwan
9.
Sci Rep ; 10(1): 12526, 2020 Jul 22.
Article in English | MEDLINE | ID: mdl-32694763

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

10.
Sci Rep ; 9(1): 4606, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30872677

ABSTRACT

The therapeutic effects of simvastatin for renal cell carcinoma (RCC) are controversial. In this study, the effects of simvastatin on the carcinogenic properties of 3-methylcholanthrene (3MC; an aryl-hydrocarbon receptor [AhR] agonist) in human renal epithelial cells (hRECs) were investigated. We exposed in vitro and in vivo models to 3MC to induce RCC onset. 3MC upregulated the epithelial-mesenchymal transition (EMT) and tumor biomarkers; the models exhibited the reciprocal expression of histone deacetylase 1 (HDAC1) and RhoA, namely increased HDAC1 and decreased RhoA expression, through hypoxia-inducible-factor (HIF)- and AhR-dependent mechanisms. In addition to inducing EMT biomarkers, 3MC decreased von Hippel-Lindau protein levels (a risk factor for RCC) and increased CD44 expression in hRECs, which were reversed by digoxin (a HIF inhibitor) and HDAC inhibitors (suberoylanilide hydroxamic acid and trichostatin A [TSA]). Simvastatin abolished the detrimental effects of 3MC by reducing HDAC1 expression, with resulting RhoA upregulation, and reactivating RhoA in vitro and in vivo. Notably, the protective effects of simvastatin were negated by an HDAC activator (ITSA) through TSA suppression. The crucial role of RhoA in RCC carcinogenesis was verified by the overexpression of constitutively active RhoA. Collectively, these results demonstrate that simvastatin restores RhoA function through HDAC1 inhibition; therefore, simvastatin might serve as adjunct therapy for RCC induced by 3MC.


Subject(s)
Epithelial Cells/drug effects , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Kidney/drug effects , Methylcholanthrene/adverse effects , Simvastatin/pharmacology , rhoA GTP-Binding Protein/metabolism , Carcinogenesis/chemically induced , Carcinogenesis/drug effects , Cells, Cultured , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/drug effects , Humans , Hyaluronan Receptors/metabolism , Hydroxamic Acids/pharmacology , Kidney/metabolism , Receptors, Aryl Hydrocarbon/metabolism
11.
Phytomedicine ; 55: 255-263, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30668437

ABSTRACT

BACKGROUND: We previously showed that 3-O-ß-D-glucopyranosyl-(3R)-hydroxybutanolide (kinsenoside), a major compound of Anoectochilus formosanus, increased lipolysis through an AMP-activated protein kinase (AMPK)-dependent pathway. PURPOSE: To extend our previous finding, we investigated the in vivo and in vitro effects of kinsenoside on lipolysis and the involvement of cyclic AMP (cAMP)-dependent protein kinase A (PKA) and AMPK in kinsenoside-mediated lipolysis. STUDY DESIGN/METHODS: Mice were fed a high-fat diet for six weeks to induce lipid deposition and then treated with 50 and 100  mg/kg kinsenoside for two weeks. The coordination of PKA and AMPK activation in lipolysis in C3H10T1/2 adipocytes was evaluated in vitro by using PKA and AMPK's corresponding inhibitors, oil-red O staining, a glycerol production assay, and Western blot analysis. RESULTS: Kinsenoside reduced body weight, fat pad mass, and hepatic lipid accumulation in obese mice, and concurrently increased the induction and activation of hormone-sensitive lipase (HSL), perilipin, adipose triglyceride lipase (ATGL), and carnitine palmitoyltransferase I (CPT1). Kinsenoside concentration-dependently increased PKA activation by increasing the phosphorylation of Ser/Thr-PKA substrates in vitro. These increases were accompanied by a reduction in fat accumulation. Using H89 and Rp-8-Br-cAMPs to inhibit PKA reduced the release of glycerol but did not alter the activation of peroxisome proliferator-activated receptor alpha or the expression of CPT1 or ATGL. By contrast, compound C, an AMPK inhibitor, inhibited CPT1 and ATGL expression in kinsenoside-treated C3H10T1/2 adipocytes. In addition, H89 caused the reactivation of AMPK downstream targets by increasing the levels of the active form of pAMPK-Thr172, suggesting that PKA negatively modulates AMPK activity. CONCLUSION: Kinsenoside increased HSL activation through PKA-mediated phosphorylation at Ser660/563 and concomitantly increased perilipin activation in lipolysis. These lipolytic effects of kinsenoside were validated using 6-Bnz-cAMPs, a PKA agonist. In this study, we demonstrated that in addition to AMPK, PKA also plays a crucial role in kinsenoside-mediated lipolysis.


Subject(s)
4-Butyrolactone/analogs & derivatives , AMP-Activated Protein Kinases/metabolism , Adipocytes/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Lipolysis/drug effects , Monosaccharides/metabolism , Plant Extracts/metabolism , Sterol Esterase/metabolism , 4-Butyrolactone/metabolism , Animals , Male , Mice , Orchidaceae/chemistry , Plant Extracts/chemistry
12.
J Renin Angiotensin Aldosterone Syst ; 19(3): 1470320318803009, 2018.
Article in English | MEDLINE | ID: mdl-30264671

ABSTRACT

INTRODUCTION: The renin-angiotensin system and epithelial-mesenchymal transition play crucial roles in the development of kidney fibrosis. The connection between the renin-angiotensin system and transforming growth factor-ß in epithelial-mesenchymal transition remains largely unknown. MATERIALS AND METHODS: We assessed oxidative stress, cytokine levels, renal morphology, profibrotic growth factor and renin-angiotensin system component expression, and cell-specific E- and N-cadherin expression in the kidneys of gerbils with streptozotocin-induced diabetes mellitus. RESULTS: Animals in the experimental group received an intraperitoneal injection of streptozotocin to induce diabetes. The diabetic gerbil kidneys presented kidney injury, which was manifested as distorted glomeruli, necrosis of tubular cells, dilated tubular lumen, and brush border loss. Additionally, the diabetic gerbil kidneys exhibited significantly higher expressions of 8-hydroxy-2'-deoxyguanosine, nuclear factor-kB, toll-like receptor 4, tumor necrosis factor-α, transforming growth factor-ß, connective tissue growth factor, α-smooth muscle actin, and N-cadherin and higher collagen deposition than did the control gerbil kidneys. Compared with the control kidneys, the diabetic gerbil kidneys exhibited significantly lower E-cadherin expression. These epithelial-mesenchymal transition characteristics were associated with an increase in renin-angiotensin system expression in the diabetic gerbils. CONCLUSIONS: We demonstrate that hyperglycemia activated the renin-angiotensin system, induced epithelial-mesenchymal transition, and contributed to kidney fibrosis in an experimental diabetes mellitus model.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Epithelial-Mesenchymal Transition , Hyperglycemia/pathology , Kidney/pathology , Renin-Angiotensin System , Actins/metabolism , Animals , Blood Glucose/metabolism , Body Weight , Cadherins/metabolism , Connective Tissue Growth Factor/metabolism , Diabetes Mellitus, Experimental/blood , Extracellular Matrix/metabolism , Fibrosis , Gerbillinae , Inflammation/pathology , Male , Oxidative Stress , Streptozocin , Transforming Growth Factor beta/metabolism
13.
Pharmacology ; 102(5-6): 262-271, 2018.
Article in English | MEDLINE | ID: mdl-30227438

ABSTRACT

We previously synthesized new tubulin inhibitors, MPT0B169 and MPT0B002, which induced growth inhibition and apoptosis in leukemia cells. However, their effects on solid tumor cells have not been determined. In this study, we investigated the effects of MPT0B169 and MPT0B002 on glioblastoma, breast, lung, and colorectal cancer (CRC) cell lines. A cell viability analysis showed that MPT0B169 and MPT0B002 were more effective in inhibiting the proliferation of COLO205 and HT29 CRC cells than U87MG and GBM8401 glioblastoma, MCF-7 and MDA-MB-231 breast cancer, and A549 lung cancer cells. MPT0B169 and MPT0B002 inhibited growth of COLO205 and HT29 cells in dose- and time-dependent manners. A colony-formation assay confirmed the growth inhibitory effects of MPT0B169 and MPT0B002 on COLO205 and HT29 cells. MPT0B169 and MPT0B002 disrupted tubulin polymerization and arrested the cell cycle at the G2/M phase, with a concomitant increase of the cyclin B1 level. MPT0B169 and MPT0B002 induced apoptosis, accompanied by induction of the intrinsic apoptotic pathway, as shown by a reduction in the caspase-9 level and increases in cleaved caspase-3 and cleaved PARP. These results suggest that MPT0B169 and MPT0B002, new tubulin inhibitors, induced growth inhibition, G2/M arrest, and apoptosis in COLO205 and HT29 cells, and they could potentially be anticancer agents for CRC cells.


Subject(s)
Colorectal Neoplasms/drug therapy , G2 Phase Cell Cycle Checkpoints/drug effects , Indoles/pharmacology , Sarcosine/analogs & derivatives , Sulfonamides/pharmacology , Tubulin Modulators/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , HT29 Cells , Humans , Sarcosine/pharmacology , Tubulin/metabolism
14.
Acta Histochem ; 120(6): 525-533, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29934127

ABSTRACT

Diabetes mellitus (DM) reduces lung function and increases the risk of asthma, chronic obstructive pulmonary disease, pneumonia, and pulmonary fibrosis. Epithelial-mesenchymal transition (EMT) plays a crucial role in the development of pulmonary fibrosis. The pathogenesis of pulmonary fibrosis in diabetes remains unknown. We investigated the effects of hyperglycemia on EMT in the lungs of gerbils with streptozotocin (STZ)-induced diabetes. Diabetic gerbils exhibited a significantly lower volume fraction of the alveolar airspace and significantly higher septal thickness, volume fraction of the alveolar wall, and lung injury scores than did nondiabetic gerbils. The percentage of 8-hydroxy-2'-deoxyguanosine-positive cells and transforming growth factor-ß-positive cells was significantly higher, the expression of E-cadherin was significantly lower, and the expression of N-cadherin was significantly higher in diabetic gerbils than in nondiabetic gerbils. These EMT characteristics were associated with a significant increase in α-smooth muscle actin (SMA) expression and collagen deposition in the lungs of diabetic gerbils. The increased α-SMA expression was co-localized with surfactant protein-C in alveolar type II cells in hyperglycemic animals. In conclusion, our study demonstrates that hyperglycemia induces EMT and contributes to lung fibrosis in an experimental DM model.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation , Hyperglycemia/metabolism , Lung/metabolism , Animals , Diabetes Mellitus, Experimental/pathology , Gerbillinae , Hyperglycemia/pathology , Lung/pathology , Male
15.
Toxicol Sci ; 160(2): 217-229, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28973641

ABSTRACT

We have previously reported that perfluorooctanesulfonate (PFOS) causes cell apoptosis in renal tubular epithelial cells (RTCs). Here, we extend our findings and provide evidence of epithelial-mesenchymal transition (EMT)-associated renal fibrosis caused by PFOS and the protection by l-carnitine. Our results demonstrate that PFOS increased the expression of EMT and renal injury biomarkers (eg, N-cadherin, vimentin, Snail, Kim1, and Lcn2). In addition, PFOS caused EMT induction through Sirt1-mediated PPARγ deacetylation and inactivation. l-carnitine reversed the EMT induction caused by PFOS and alleviated PFOS-mediated increases in cell migration by reactivating PPARγ through the inhibition of Sirt1 activity. The critical role of Sirt1 in this process was validated by using Sirt1 overexpression, resveratrol (a pharmacologic activator of Sirt1), nicotinamide (a Sirt1 inhibitor) and siSirt1. Nicotinamide and siSirt1, but not Sirt1 overexpression and resveratrol, alleviated PFOS-mediated EMT induction, suggesting that increased Sirt1 activity contributed to the alterations. Furthermore, through PPARγ overexpression and pharmacologic interventions, we validated the crucial role of increased PPARγ deacetylation caused by aberrant increased Sirt1 activity in RTC transformation. Similar to PPARγ overexpression, rosiglitazone (a PPARγ agonist) alleviated the effects of PFOS on the EMT-related features, whereas GW9662 (a PPARγ antagonist) mimicked the effects. The protective effect of l-carnitine was also verified in a mouse model of chronic PFOS exposure, in which decreased EMT biomarker levels and renal fibrosis by l-carnitine were observed in Western blot and histological analyses. Accordingly, l-carnitine alleviated EMT-associated renal fibrosis caused by PFOS through a Sirt1- and PPARγ-dependent mechanism.


Subject(s)
Alkanesulfonic Acids/toxicity , Carnitine/pharmacology , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Fluorocarbons/toxicity , Kidney Diseases/prevention & control , Kidney Tubules/drug effects , PPAR gamma/metabolism , Protective Agents/pharmacology , Sirtuin 1/metabolism , Acetylation , Animals , Cell Line , Cell Movement/drug effects , Cytoprotection , Disease Models, Animal , Dose-Response Relationship, Drug , Epithelial Cells/enzymology , Epithelial Cells/pathology , Fibrosis , Kidney Diseases/enzymology , Kidney Diseases/pathology , Kidney Tubules/enzymology , Kidney Tubules/pathology , Male , Mice, Inbred BALB C , PPAR gamma/genetics , Rats , Signal Transduction/drug effects , Sirtuin 1/genetics , Time Factors , Transfection
16.
PLoS One ; 12(8): e0183187, 2017.
Article in English | MEDLINE | ID: mdl-28806401

ABSTRACT

Growth differentiation factor 15 (GDF15) is a strong predictor of cardiovascular events and mortality in individuals with or without cardiovascular diseases. Single nucleotide polymorphisms (SNPs) in microRNA (miRNA) target sites, also known as miRSNPs, are known to enhance or weaken miRNA-mRNA interactions and have been linked to diseases such as cardiovascular disease and cancer. In this study, we aimed to elucidate the functional significance of the miRSNP rs1054564 in regulating GDF15 levels. Two rs1054564-containing binding sites for hsa-miR-873-5p and hsa-miR-1233-3p were identified in the 3' untranslated region (UTR) of the GDF15 transcript using bioinformatics tools. Their activities were further characterized by in vitro reporter assays. Bioinformatics prediction suggested that miRNA binding sites harboring the rs1054564-G allele had lower free energies than those with the C allele and therefore were better targets with higher affinities for both hsa-miR-873-5p and hsa-miR-1233-3p. Reporter assays showed that luciferase activity was significantly decreased by rs1054564-G-containing 3' UTRs for both miRNAs (P < 0.05) and was restored by miRNA inhibitors. Comparing the fold suppression of the two miRNAs, only that of hsa-miR-1233-3p showed significant changes between the rs1054564-G- and C-containing 3' UTRs (P = 0.034). In addition, western blots showed that transfection of both miRNA mimics significantly decreased endogenous GDF15 expression in a melanoma cell line (P < 0.05). Taken together, our findings demonstrate that GDF15 is a target of hsa-miR-873-5p and hsa-miR-1233-3p and that the rs1054564-C allele partially abolishes hsa-miR-1233-3p-mediated translational suppression of GDF15. These results suggest that rs1054564 confers allele-specific translational repression of GDF15 via hsa-miR-1233-3p. Our work thus provides biological insight into the previously reported clinical association between rs1054564 and plasma GDF15 levels.


Subject(s)
3' Untranslated Regions/genetics , Alleles , Growth Differentiation Factor 15/genetics , MicroRNAs/metabolism , Polymorphism, Single Nucleotide/genetics , Protein Biosynthesis/genetics , Base Sequence , Binding Sites/genetics , Cell Line, Tumor , Computer Simulation , Growth Differentiation Factor 15/metabolism , HEK293 Cells , Humans , MicroRNAs/chemistry , MicroRNAs/genetics , Nucleic Acid Conformation
17.
J Cell Physiol ; 232(5): 1020-1029, 2017 05.
Article in English | MEDLINE | ID: mdl-27442426

ABSTRACT

Endothelial nitric oxide synthase (eNOS) modulates vascular blood pressure and is predominantly expressed in endothelial cells and activated through the protein kinase B (Akt/PKB)-dependent pathway. We previously reported that 3-methylcholanthrene (3MC) activates the aryl hydrocarbon receptor (AhR) and reduces PI3K/Akt phosphorylation. This study investigated the mechanism underlying the downregulatory effects of 3-MC on nitric oxide (NO) production occurring through the AhR/RhoA/Akt-mediated mechanism. The mechanism underlying the effects of 3-MC on eNOS activity and blood pressure was examined in vitro and in vivo through genetic and pharmacological approaches. Results indicated that 3-MC modified heat shock protein 90 (HSP90), caveolin-1, dynein, and eNOS mRNA and protein expression through the AhR/RhoA-dependent mechanism in mouse cerebral vascular endothelial cells (MCVECs) and that 3-MC reduced eNOS phosphorylation through the AhR/RhoA-mediated inactivation of Akt1. The upregulation of dynein expression was associated with decreased eNOS dimer formation (eNOS dimer; an activated form of the enzyme). Coimmunoprecipitation assay results indicated that 3-MC significantly reduced the interaction between eNOS and its regulatory proteins, including Akt1 and HSP90, but increased the interaction between eNOS and caveolin-1. Immunofluorescence and Western blot analysis revealed that 3-MC reduced the amount of membrane-bound activated eNOS, and a modified Griess assay revealed that 3-MC concomitantly reduced NO production. However, simvastatin reduced 3-MC-mediated murine hypertension. Our study results indicate that AhR, RhoA, and eNOS have major roles in blood pressure regulation. Statin intervention might provide a potential therapeutic approach for reducing hypertension caused by 3-MC. J. Cell. Physiol. 232: 1020-1029, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Hypertension/enzymology , Methylcholanthrene/pharmacology , Nitric Oxide Synthase Type III/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Cerebrum/blood supply , Down-Regulation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Hypertension/pathology , Mice , Models, Biological , Nitric Oxide/metabolism , Nitroprusside/pharmacology , Nitroprusside/therapeutic use , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Simvastatin/pharmacology , Simvastatin/therapeutic use , Time Factors , rhoA GTP-Binding Protein/metabolism
18.
PLoS One ; 11(5): e0155190, 2016.
Article in English | MEDLINE | ID: mdl-27171144

ABSTRACT

Perfluorinated chemicals (PFCs) are ubiquitously distributed in the environments including stainless pan-coating, raincoat, fire extinguisher, and semiconductor products. The PPAR family has been shown to contribute to the toxic effects of PFCs in thymus, immune and excretory systems. Herein, we demonstrated that perfluorooctanesulfonate (PFOS) caused cell apoptosis through increasing ratio of Bcl-xS/xL, cytosolic cytochrome C, and caspase 3 activation in renal tubular cells (RTCs). In addition, PFOS increased transcription of inflammatory cytokines (i.e., TNFα, ICAM1, and MCP1) by NFκB activation. Conversely, PFOS reduced the mRNA levels of antioxidative enzymes, such as glutathione peroxidase, catalase, and superoxide dismutase, as a result of reduced PPARγ transactivational activity by using reporter and chromatin immuoprecipitation (ChIP) assays. PFOS reduced the protein interaction between PPARγ and PPARγ coactivator-1 alpha (PGC1α) by PPARγ deacetylation through Sirt1 upregulation, of which the binding of PPARγ and PGC1α to a peroxisome proliferator response element (PPRE) in the promoter regions of these antioxidative enzymes was alleviated in the ChIP assay. Furthermore, Sirt1 also deacetylated p53 and then increased the binding of p53 to Bax, resulting in increased cytosolic cytochrome C. The effect of PPARγ inactivation by PFOS was validated using the PPARγ antagonist GW9662, whereas the adverse effects of PFOS were prevented by PPARγ overexpression and activators, rosiglitozone and L-carnitine, in RTCs. The in vitro finding of protective effect of L-carnitine was substantiated in vivo using Balb/c mice model subjected to PFOS challenge. Altogether, we provide in vivo and in vitro evidence for the protective mechanism of L-carnitine in eliminating PFOS-mediated renal injury, at least partially, through PPARγ activation.


Subject(s)
Alkanesulfonic Acids/toxicity , Apoptosis/drug effects , Fluorocarbons/toxicity , Kidney Tubules/pathology , PPAR gamma/metabolism , Acetylation/drug effects , Animals , Antioxidants/metabolism , Carnitine/pharmacology , Cell Line , Cell Proliferation/drug effects , Cytochromes c/metabolism , Cytosol/metabolism , Genes, Reporter , Inflammation/pathology , Kidney Function Tests , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Kidney Tubules/physiopathology , Male , Mice, Inbred BALB C , Oxidation-Reduction/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protective Agents/pharmacology , Protein Binding/drug effects , Rats , Response Elements/genetics , Rosiglitazone , Signal Transduction/drug effects , Sirtuin 1/metabolism , Thiazolidinediones/pharmacology , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
19.
Phytomedicine ; 22(6): 641-7, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-26055129

ABSTRACT

BACKGROUND: Currently, more than one-third of the global population is overweight or obese, which is a risk factor for major causes of death including cardiovascular disease, numerous cancers, and diabetes. Kinsenoside, a major active component of Anoectochilus formosanus exhibits antihyperglycemic, antihyperliposis, and hepatoprotective effects and can be used to prevent and manage obesity. PURPOSE: This study examined the catabolic effects of kinsenoside on lipolysis in adipocytes transformed from C3H10T1/2 cells. STUDY DESIGN/METHODS: The lipolytic effect of kinsenoside in C3H10T1/2 adipocytes was evaluated by oil-red O staining and glycerol production. The underlying mechanisms were assessed by Western blots, chromatin immunoprecipitation (IP), Co-IP, EMSA and siRNAs verification. RESULTS: We demonstrated that kinsenoside increased both adipose triglyceride lipase (ATGL)-mediated lipolysis, which was upregulated by AMP-activated protein kinase (AMPK) activation, and the hydrolysis of triglycerides to glycerol and fatty acids that require transportation into mitochondria for further ß-oxidation. We also demonstrated that kinsenoside increased the phosphorylation of peroxisome proliferator-activated receptor alpha (PPARα) and CRE-binding protein (CREB), and the protein levels of silent information regulator T1 (SIRT1), peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) and carnitine palmitoyltransferase I (CPT1) through an AMPK-dependent mechanism. SIRT1 deacetylated PGC-1α, facilitating AMPK-mediated PGC-1α phosphorylation and increasing the interaction of PPARα with its coactivator, PGC-1α. This interaction elevated the expression of CPT1, a shuttle for the mitochondrial transport of fatty acids, in kinsenoside-treated cells. In addition, AMPK-phosphorylation-mediated CREB activation caused kinsenoside-mediated PGC-1α upregulation. CONCLUSION: AMPK activation not only elevated ATGL expression for lipolysis but also induced CPT1 expression for further mitochondrial translocation of fatty acids. The results suggested that the mechanism underlying the catabolic effects of kinsenoside on lipolysis and increased CPT1 induction was mediated through an AMPK-dependent pathway.


Subject(s)
4-Butyrolactone/analogs & derivatives , AMP-Activated Protein Kinases/metabolism , Adipocytes/drug effects , Lipolysis , Monosaccharides/pharmacology , PPAR alpha/metabolism , 4-Butyrolactone/pharmacology , Adipocytes/metabolism , Animals , Carnitine O-Palmitoyltransferase/metabolism , Cell Line , Fatty Acids/metabolism , Glycerol/metabolism , Mice , Mitochondria/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Signal Transduction , Sirtuin 1 , Transcription Factors/metabolism
20.
PLoS One ; 10(4): e0122664, 2015.
Article in English | MEDLINE | ID: mdl-25875811

ABSTRACT

OBJECTIVE: Previous genome-wide association studies have indicated an association between CDH13 genotypes and adiponectin levels. In this study, we used mediation analysis to assess the statistical association between CDH13 locus variants and adiponectin levels, metabolic syndrome, and related metabolic phenotypes. METHODS AND RESULTS: A sample population of 530 Taiwanese participants was enrolled. Four CDH13 gene variants in the promoter and intron 1 regions were genotyped. After adjustment for clinical covariates, the CDH13 genotypes/haplotypes exhibited an association with the adiponectin levels (lowest P = 1.95 × 10-11 for rs4783244 and lowest P = 3.78 × 10-13 for haplotype ATTT). Significant correlations were observed between the adiponectin levels and the various metabolic syndrome-related phenotypes (all P ≤ 0.005). After further adjustment for the adiponectin levels, participants with a minor allele of rs12051272 revealed a considerable association with a more favorable metabolic profile, including higher insulin sensitivity, high-density lipoprotein cholesterol levels, lower diastolic blood pressure, circulating levels of fasting plasma glucose, and triglycerides, and as a lower risk of metabolic syndrome (all P < 0.05). The mediation analysis further revealed a suppression effect of the adiponectin levels on the association between CDH13 genotypes and metabolic syndrome and its related phenotypes (Sobel test; all P < 0.001). CONCLUSION: The genetic polymorphisms at the CDH13 locus independently affect the adiponectin levels, whereas the adiponectin levels exhibit a suppressive effect on the association between CDH13 locus variants and various metabolic phenotypes and metabolic syndrome. In addition, these results provide further evidence of the association between the CDH13 gene variants and the risks of metabolic syndrome and atherosclerotic cardiovascular disease.


Subject(s)
Adiponectin/genetics , Cadherins/genetics , Glucose/metabolism , Metabolic Syndrome/genetics , Adiponectin/blood , Adult , Alleles , Cadherins/blood , Cholesterol/genetics , Cholesterol/metabolism , Female , Genome-Wide Association Study , Genotype , Haplotypes , Humans , Insulin/genetics , Insulin/metabolism , Male , Metabolic Syndrome/blood , Metabolic Syndrome/metabolism , Middle Aged , Polymorphism, Single Nucleotide , Taiwan
SELECTION OF CITATIONS
SEARCH DETAIL
...