Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Front Oncol ; 14: 1361152, 2024.
Article in English | MEDLINE | ID: mdl-38515566

ABSTRACT

Background: Prostate cancer (PCa) is the second most common solid cancer among men worldwide and the fifth leading cause of cancer-related deaths in men. Sulforaphane (SFN), an isothiocyanate compound, has been shown to exert inhibitory effects on a variety of cancers. However, the biological function of SFN in PCa has not been fully elucidated. The objective of this study was conducted to further investigate the possible underlying mechanism of SFN in PCa using in vitro cell culture and in vivo tumor model experiments. Methods: Cell viability, migration, invasion, and apoptosis were analyzed by Cell Counting Kit-8 (CCK-8), wound healing assay, transwell assay, or flow cytometry. Expression of microRNA (miR)-3919 was detected by quantitative real-time polymerase chain reaction (qRT-PCR) or in situ hybridization assay. Xenograft assay was conducted to validated the antitumor effect of miR-3919. The targeting relationship between miR-3919 and DJ-1 was verified by dual-luciferase reporter assay. The level of DJ-1was measured by qRT-PCR or western blotting (WB). Results: In the present study, SFN downregulated mRNA and protein expression of DJ-1, an oncogenic gene. Small RNA sequencing analysis and dual-luciferase reporter assay confirmed that microRNA (miR)-3919 directly targeted DJ-1 to inhibition its expression. Furthermore, miR-3919 overexpression impeded viability, migration, and invasion and promoted apoptosis of PCa cells. Tumor growth in nude mice was also inhibited by miR-3919 overexpression, and miR-3919 expression in PCa tissues was lower than that in peritumoral tissues in an in situ hybridization assay. Transfection with miR-3919 inhibitors partially reversed the effects of SFN on cell viability, migration, invasion, and apoptosis. Conclusion: Overall, the miR-3919/DJ-1 axis may be involved in the effects of SFN on the malignant biological behavior of PCa cells, which might be a new therapeutic target in PCa.

2.
Sci Total Environ ; 883: 163662, 2023 Jul 20.
Article in English | MEDLINE | ID: mdl-37100122

ABSTRACT

The migration and transformations of Cd and As in soil are different, so it is difficult to simultaneously control them. In this study, an organo-mineral complex (OMC) material was prepared using modified palygorskite and chicken manure, the Cd and As adsorption capacities and mechanism of the OMC were explored, and the response of the crop to the OMC was clarified. The results show that the maximum Cd and As adsorption capacities of the OMC under pH values of 6-8 are 12.19 mg·g-1 and 5.07 mg·g-1, respectively. In the OMC system, the modified palygorskite contributed more to the adsorption of the heavy metals than the organic matter. Cd2+ may form CdCO3 and CdFe2O4, and AsO2- may form FeAsO4, As2O3, and As2O5 on the surfaces of the modified palygorskite. Organic functional groups such as hydroxyl, imino, and benzaldehyde groups can participate in the adsorption of Cd and As. The Fe species and carbon vacancy in the OMC system promote the conversion of As3+ into As5+. A laboratory experiment was conducted to compare five commercial remediation agents with OMC. Planting Brassica campestris in the OMC remediated soil with excessive contamination increased the crop biomass and decreased the Cd and As accumulation sufficiently to meet the current national food safety standards. This study emphasizes the effectiveness of OMC in preventing the migration of Cd and As into crops while promoting crop growth, which can provide a feasible soil management strategy for CdAs co-contaminated farmland soil.

3.
Front Cell Dev Biol ; 9: 666303, 2021.
Article in English | MEDLINE | ID: mdl-34631693

ABSTRACT

The WNT signaling pathway plays a crucial role in oviduct/fallopian development. However, the specific physiological processes regulated by the WNT pathway in the fallopian/oviduct function remain obscure. Benefiting from the Lgr4 knockout mouse model, we report the regulation of oviduct epithelial secretion by LGR4. Specifically, the loss of Lgr4 altered the mouse oviduct size and weight, severely reduced the number of oviductal epithelial cells, and ultimately impaired the epithelial secretion. These alterations were mediated by a failure of CTNNB1 protein accumulation in the oviductal epithelial cytoplasm, by the modulation of WNT pathways, and subsequently by a profound change of the gene expression profile of epithelial cells. In addition, selective activation of the WNT pathway triggered the expression of steroidogenic genes, like Cyp11a1 and 3ß-Hsd1, through the activation of the transcriptional factor NR5A2 in an oviduct primary cell culture system. As demonstrated, the LGR4 protein modulates a WNT-NR5A2 signaling cascade facilitating epithelial secretory cell maturation and steroidogenesis to safeguard oviduct development and function in mice.

4.
J Assist Reprod Genet ; 38(12): 3175-3193, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34664184

ABSTRACT

Oxidative stress is one of the major causes leading to male infertility including asthenozoospermia. Hydrogen sulfide (H2S) has been widely recognized to be a potent antioxidant whose role is partially implemented by protein S-sulfhydration. However, protein S-sulfhydration has not been reported in germ cells. Therefore, we investigated whether asthenozoospermia could be associated with sperm protein S-sulfhydration. S-sulfhydrated proteins in human sperm were enriched via biotin-switch assay and analyzed using LC-MS/MS spectrometry. Two hundred forty-four S-sulfhydrated proteins were identified. Importantly, we validated that sperm histones H3.1 and H3.3 were the S-sulfhydrated proteins. Their S-sulfhydrated amino acid residue was Cysteine111. Abundances of S-sulfhydrated H3 (sH3) and S-sulfhydrated H3.3 (sH3.3) were significantly down-regulated in asthenozoospermic sperm, compared with the fertile controls, and were significantly correlated with progressive motility. Retinoic acid (RA) up-regulated level of sH3.3 in primary round spermatids and the C18-4 cells (a mouse spermatogonial stem cell line). Overexpression of the mutant H3.3 (Cysteine111 was replaced with serine) affected expression of 759 genes and raised growth rate of C18-4 cells. For the first time, S-sulfhydration H3 and H3.3 were demonstrated in the present study. Our results highlight that aberrant S-sulfhydration of H3 is a new pathophysiological basis in male infertility.


Subject(s)
Asthenozoospermia/physiopathology , Cysteine/metabolism , Histones/metabolism , Spermatozoa/metabolism , Sulfhydryl Compounds/metabolism , Amino Acid Sequence , Animals , Biotin/metabolism , Gene Expression Regulation , Humans , Hydrogen Sulfide/metabolism , Infertility/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation , Protein Processing, Post-Translational , Spermatogenesis , Sulfides/metabolism
5.
Life Sci ; 278: 119570, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-33964295

ABSTRACT

AIMS: Increasing evidence has shown that hormone secretion is regulated by endocytosis. Eps15 homology domain-containing protein 3 (EHD3) is an endocytic-trafficking regulatory protein, but whether EHD3 is associated with testosterone secretion is not clear. This work aims to explore the role of EHD3 in testosterone synthesis. MAIN METHODS: Testosterone concentration was determined by ELISA. The effects of EHD3 on endocytosis were assessed by exosomes tracing assay and Immunofluorescence. Targeting relationship between EHD3 and NR5A1 was verified by chromatin immunoprecipitation (ChIP) and dual luciferase reporter gene assay in Leydig cells. For in vivo assessments, conditional NR5A1 knockout mouse model was established with CRISPR/Cas9 gene targeting technology. KEY FINDINGS: EHD3 overexpression significantly increased the concentration of testosterone. EHD3 knockdown markedly decreased testosterone synthesis by reducing endocytosis. The activity of the EHD3 promoter was positively regulated by NR5A1, which occupied the conserved sequence "AGGTCA" in the EHD3 promoter. Furthermore, mice with a Leydig cell-specific conditional NR5A1 knockout displayed the blunted levels of EHD3 and clathrin (a key factor for endocytosis), and serum testosterone concentration compared with NR5A1f/f mice. SIGNIFICANCE: This study suggests a potential molecular mechanism of testosterone synthesis to fully understand male reproductive health.


Subject(s)
Carrier Proteins/metabolism , Endocytosis , Exosomes/metabolism , Gene Expression Regulation , Steroidogenic Factor 1/metabolism , Testosterone/metabolism , Animals , CRISPR-Cas Systems , Carrier Proteins/genetics , Chromatin Immunoprecipitation , Female , Leydig Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasmids/metabolism , Promoter Regions, Genetic , RNA, Small Interfering/metabolism , Steroidogenic Factor 1/genetics , Testosterone/pharmacology
6.
Sci Rep ; 6: 28908, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27349344

ABSTRACT

Chronic cerebral hypoperfusion (CCH) is one of the most common causes of vascular dementia (VaD) and is recognised as an etiological factor in the development of Alzheimer's disease (AD). CCH can induce severe cognitive deficits, as assessed by the water maze task, along with neuronal loss in the hippocampus. However, there are currently no effective, approved pharmacological treatments available for VaD. In the present study, we created a rat model of CCH using bilateral common carotid artery occlusion and found that (-)-SCR1693, a novel compound, prevented rats from developing memory deficits and neuronal damage in the hippocampus by rectifying cholinergic dysfunction and decreasing the accumulation of the phospho-tau protein. These results strongly suggest that (-)-SCR1693 has therapeutic potential for the treatment of CCH-induced VaD.


Subject(s)
Cerebrovascular Disorders/physiopathology , Hippocampus/drug effects , Memory Disorders/prevention & control , Tacrine/analogs & derivatives , Animals , Arterial Occlusive Diseases/metabolism , Arterial Occlusive Diseases/physiopathology , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/physiopathology , Cerebrovascular Disorders/metabolism , Chronic Disease , Dementia, Vascular/metabolism , Dementia, Vascular/physiopathology , Dementia, Vascular/prevention & control , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Maze Learning/drug effects , Memory Disorders/metabolism , Memory Disorders/physiopathology , Neurons/metabolism , Protective Agents/pharmacology , Rats, Sprague-Dawley , Tacrine/pharmacology , tau Proteins/metabolism
7.
Sci Rep ; 6: 24233, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27052887

ABSTRACT

The purpose of this study was to characterize a novel compound, 4-[2-(dimethylamino)-1-(1-hydroxycyclohexyl) ethyl] phenyl 3-nitrophenyl ether, designated LPM580153. We used several well-validated animal models of depression to assess the antidepressant-like activity of LPM580153, followed by a neurotransmitter uptake assay and a corticosterone-induced cell injury model to explore its mechanism of action. In mice, LPM580153 reduced immobility time in the tail suspension test, and in rats subjected to chronic unpredictable mild stress it reversed reductions in body weight gain and ameliorated anhedonia. The neurotransmitter uptake assay results demonstrated that LPM580153 inhibited the uptake of serotonin, norepinephrine and dopamine. Furthermore, LPM580153 protected the SH-SY5Y cells against the cytotoxic activity of corticosterone, an action that might be related to the role of LPM580153 in increasing the protein levels of BDNF, p-ERK1/2, p-AKT, p-CREB and p-mTOR. Together, these findings indicate that LPM580153 is a novel triple reuptake inhibitor with robust antidepressant-like effects.


Subject(s)
Anhedonia/drug effects , Antidepressive Agents/pharmacology , Cyclohexanols/pharmacology , Depression/prevention & control , Neurotransmitter Uptake Inhibitors/pharmacology , Phenethylamines/pharmacology , Animals , Antidepressive Agents/chemistry , Biological Transport/drug effects , Blotting, Western , Brain-Derived Neurotrophic Factor/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Corticosterone/pharmacology , Cyclohexanols/chemistry , Depression/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopamine/pharmacokinetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Male , Mice , Molecular Structure , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurotransmitter Uptake Inhibitors/chemistry , Norepinephrine/metabolism , Norepinephrine/pharmacokinetics , Phenethylamines/chemistry , Rats, Sprague-Dawley , Serotonin/metabolism , Serotonin/pharmacokinetics , Weight Gain/drug effects
8.
Sci Rep ; 4: 5632, 2014 Jul 09.
Article in English | MEDLINE | ID: mdl-25005489

ABSTRACT

Adhyperforin is a novel constituent of Hypericum perforatum L., but its antidepressant-like activity remains unclear. To explore that, several well-validated animal models of depression as well as neurotransmitter reuptake and transporter binding assays were conducted. The results showed adhyperforin could reduce the immobility time of mice in the forced swimming test and tail suspension assay, antagonize the behaviors induced by reserpine, and have no effect on locomotor activity. Furthermore, following establishment of a chronic unpredictable mild stress model, adhyperforin increased the number of crossings and rearings in rats in the open field test and increased the sucrose consumption. Finally, adhyperforin inhibited uptake of serotonin, norepinephrine, and dopamine, and displayed robust binding affinities for the serotonin and norepinephrine transporters. Overall, the current study provides the first evidence that adhyperforin is a novel, active ingredient of Hypericum perforatum L. with robust antidepressant-like activity.


Subject(s)
Antidepressive Agents/pharmacology , Depression/drug therapy , Hypericum/chemistry , Plant Extracts/pharmacology , Terpenes/pharmacology , Animals , Behavior, Animal/drug effects , Dopamine/metabolism , Hindlimb Suspension/methods , Male , Mice , Motor Activity/drug effects , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley , Reserpine/pharmacology , Serotonin/metabolism , Swimming/physiology
9.
Sci Rep ; 4: 4986, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24832194

ABSTRACT

The clinical use of cisplatin was severely limited by its associated nephrotoxicity. In this study, we investigated whether the pseudoginsenoside F11 had protective effects against cisplatin-induced nephrotoxicity. To clarify it, one in vivo model of cisplatin-induced acute renal failure was performed. The results showed that pretreatment with F11 reduced cisplatin-elevated blood urea nitrogen and creatinine levels, as well as ameliorated the histophathological damage. Further studies showed that F11 could suppress P53 activation, inverse the ratio of Bax/Bcl2 and the anti-oxidative and free radical levels induced by cisplatin, which in turn inhibited tubular cell apoptosis. Importantly, F11 enhanced rather than inhibited the anti-tumor activity of cispaltin in murine melanoma and Lewis lung cancer xenograft tumor models. Our findings suggested that administering F11 with cisplatin might alleviate the associated nephrotoxicity without compromising its therapeutic efficiency. This finding provides a novel potential strategy in the clinical treatment of cancer.


Subject(s)
Acute Kidney Injury/chemically induced , Antineoplastic Agents/pharmacology , Cisplatin/adverse effects , Ginsenosides/pharmacology , Kidney Tubules/drug effects , Acute Kidney Injury/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/metabolism , Kidney Tubules/metabolism , Male , Melanoma/drug therapy , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Xenograft Model Antitumor Assays/methods
10.
PLoS One ; 9(4): e95777, 2014.
Article in English | MEDLINE | ID: mdl-24755839

ABSTRACT

The neuroprotective activity of pyruvate has been confirmed in previous in vivo and in vitro studies. Here, we report a novel mechanism that pyruvate prevents SH-SY5Y cells from glutamate excitotoxicity by regulating death-associated protein kinase 1 (DAPK1) protein complex. Our results showed pyruvate regulated DAPK1 protein complex to protect cells by two ways. First, pyruvate induced the dissociation of DAPK1 with NMDA receptors. The disruption of the DAPK1-NMDA receptors complex resulted in a decrease in NMDA receptors phosphorylation. Then the glutamate-stimulated Ca2+ influx was inhibited and intracellular Ca2+ overload was alleviated, which blocked the release of cytochrome c and cell death. In addition, increased Bcl-xL induced by pyruvate regulated Bax/Bak dependent death by inhibiting the release of cytochrome c from the mitochondrial inter-membrane space into the cytosol. As a result, the cytochrome c-initiated caspase cascade, including caspase-3 and caspase-9, was inhibited. Second, pyruvate promoted the association between DAPK1 and Beclin-1, which resulted in autophagy activation. The autophagy inhibitor 3-methyladenine reversed the protection afforded by pyruvate. Furthermore, the attenuation of mitochondrial damage induced by pyruvate was partly reduced by 3-methyladenine. This suggested autophagy mediated pyruvate protection by preventing mitochondrial damage. Taken together, pyruvate protects cells from glutamate excitotoxicity by regulating DAPK1 complexes, both through dissociation of DAPK1 from NMDA receptors and association of DAPK1 with Beclin-1. They go forward to protect cells by attenuating Ca2+ overload and activating autophagy. Finally, a convergence of the two ways protects mitochondria from glutamate excitotoxicity, which leads to cell survival.


Subject(s)
Death-Associated Protein Kinases/metabolism , Excitatory Amino Acid Agonists/toxicity , Glutamic Acid/toxicity , Neuroprotective Agents/pharmacology , Pyruvic Acid/pharmacology , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Beclin-1 , Calcium/metabolism , Caspase 3/metabolism , Cell Line , Cytochromes c/metabolism , Humans , Membrane Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Phosphorylation , Protein Binding , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
11.
Article in English | MEDLINE | ID: mdl-23956772

ABSTRACT

The use of doxorubicin (Dox) was severely constrained by dose-dependent side effects, which might be attenuated by combining a "sensitizer" to decrease its cumulative dosage. In this study, it was investigated whether ocotillol could enhance the antiproliferation activity of Dox. MTT assays and xenograft tumor model were firstly conducted to evaluate the effect of ocotillol on the antitumor activity of Dox. Flow cytometry and Hoechst staining assays were then performed to assess cell apoptosis. Western blot and real-time PCR were finally used to detect the expression of p53 and its target genes. Our results showed ocotillol to enhance Dox-induced cell death in p53 wild-type cancer cells. Compared with Dox alone, Dox with ocotillol (Dox-O) could induce much more cell apoptosis and activate p53 to a much greater degree, which in turn markedly increased expression of proapoptosis genes. The enhanced cytotoxic activity was partially blocked by pifithrin- α , which might be through attenuating the increased apoptosis. Furthermore, ocotillol significantly increased the antitumor activity of Dox in A549 xenograft tumor in nude mice. These findings indicated that ocotillol could potentiate the cytotoxic effect of Dox through p53-dependent apoptosis and suggested that coadministration of ocotillol with Dox might be a potential therapeutic strategy.

12.
Int J Nanomedicine ; 8: 1-11, 2013.
Article in English | MEDLINE | ID: mdl-23293515

ABSTRACT

To improve the pharmacokinetics and stability of recombinant human erythropoietin (rhEPO), rhEPO was successfully formulated into poly(ethylene glycol)-poly(d,l-lactide) (PEG-PLA) di-block copolymeric micelles at diameters ranging from 60 to 200 nm with narrow polydispersity indices (PDIs; PDI < 0.3) and trace amount of protein aggregation. The zeta potential of the spherical micelles was in the range of -3.78 to 4.65 mV and the highest encapsulation efficiency of rhEPO in the PEG-PLA micelles was about 80%. In vitro release profiles indicated that the stability of rhEPO in the micelles was improved significantly and only a trace amount of aggregate was found. Pharmacokinetic studies in rats showed highly enhanced plasma retention time of the rhEPO-loaded PEG-PLA micelles in comparison with the native rhEPO group. Increased hemoglobin concentrations were also found in the rat study. Native polyacrylamide gel electrophoresis results demonstrated that rhEPO was successfully encapsulated into the micelles, which was stable in phosphate buffered saline with different pHs and concentrations of NaCl. Therefore, PEG-PLA micelles can be a potential protein drug delivery system.


Subject(s)
Erythropoietin/administration & dosage , Micelles , Nanocapsules/chemistry , Polyethylene Glycols/chemistry , Analysis of Variance , Animals , Area Under Curve , Drug Stability , Erythropoietin/chemistry , Erythropoietin/pharmacokinetics , Female , Hemoglobins/analysis , Hemoglobins/drug effects , Humans , Nanocapsules/administration & dosage , Particle Size , Polyethylene Glycols/administration & dosage , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Sodium Chloride
SELECTION OF CITATIONS
SEARCH DETAIL
...