Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Glia ; 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38829008

ABSTRACT

As one of the top causes of blindness worldwide, glaucoma leads to diverse optic neuropathies such as degeneration of retinal ganglion cells (RGCs). It is widely accepted that the level of intraocular pressure (IOP) is a major risk factor in human glaucoma, and reduction of IOP level is the principally most well-known method to prevent cell death of RGCs. However, clinical studies show that lowering IOP fails to prevent RGC degeneration in the progression of glaucoma. Thus, a comprehensive understanding of glaucoma pathological process is required for developing new therapeutic strategies. In this study, we provide functional and histological evidence showing that optic nerve defects occurred before retina damage in an ocular hypertension glaucoma mouse model, in which oligodendroglial lineage cells were responsible for the subsequent neuropathology. By treatment with clemastine, an Food and Drug Administration (FDA)-approved first-generation antihistamine medicine, we demonstrate that the optic nerve and retina damages were attenuated via promoting oligodendrocyte precursor cell (OPC) differentiation and enhancing remyelination. Taken together, our results reveal the timeline of the optic neuropathies in glaucoma and highlight the potential role of oligodendroglial lineage cells playing in its treatment. Clemastine may be used in future clinical applications for demyelination-associated glaucoma.

2.
Small ; 18(5): e2105589, 2022 02.
Article in English | MEDLINE | ID: mdl-34908234

ABSTRACT

Polyetheretherketone (PEEK) is widely used in orthopedic and craniomaxillofacial surgeries as it exhibits excellent biocompatibility, mechanical property, and chemical stability. However, its clinical application is limited by the biological inertness of PEEK. Numerous efforts have been made to improve the bioactivity of this polymer over the years. However, modification methods that can not only promote osteogenesis but also maintain excellent properties are still limited. Hence, a facile hot die formation technique is developed for establishing patterned nanorod arrays on the PEEK surface in situ. This method can maintain the excellent properties of PEEK and can be used in implantation as it can facilitate osteogenic activity in the absence of any organic/inorganic differentiation-inducing factors. PEEK with 200-nm patterned nanorod arrays on the surface exhibits excellent osteogenic properties. This result is obtained by assessing the osteogenic differentiation properties of rat adipose-derived stem cells at the gene and protein levels in vitro. In vivo experimental results reveal that the surface-modified cylindrical PEEK 200 implants present with excellent osseointegration properties. Moreover, they can tightly bind with the surrounding bone tissue. A practical method for manufacturing single-component PEEK implants with excellent osseointegration properties is reported, and the materials can be possibly used as orthopedic implants.


Subject(s)
Nanotubes , Osseointegration , Animals , Benzophenones , Osteogenesis , Polyethylene Glycols/chemistry , Polymers , Rats , Surface Properties
3.
Small ; 18(26): e2202485, 2022 07.
Article in English | MEDLINE | ID: mdl-35633288

ABSTRACT

Mesenchymal stem cells (MSCs) have been recognized as one of the most promising pharmaceutical multipotent cells, and a key step for their wide application is to safely and efficiently regulate their activities. Various methods have been proposed to regulate the directional differentiation of MSCs during tissue regeneration, such as nanoparticles and metal ions. Herein, nanoscale zeolitic imidazolate framework-8 (ZIF-8), a Zn-based metal-organic framework, is modified to direct MSCs toward an osteoblast lineage. Specifically, ZIF-8 nanoparticles are encapsulated using stem cell membranes (SCMs) to mimic natural molecules and improve the biocompatibility and targeted ability toward MSCs. SCM/ZIF-8 nanoparticles adjust the sustained release of Zn2+ , and promote their specific internalization toward MSCs. The internalized SCM/ZIF-8 nanoparticles show excellent biocompatibility, and increase MSCs' osteogenic potentials. Moreover, RNA-sequencing results elucidate that the activated cyclic adenosine 3,5-monophosphate (cAMP)-PKA-CREB signaling pathway can be dominant in accelerating osteogenic differentiation. In vivo, SCM/ZIF-8 nanoparticles greatly promote the formation of new bone tissue in the femoral bone defect detected by 3D micro-CT, hematoxylin and eosin staining, and Masson staining after 4 weeks. Overall, the SCM-derived ZIF-8 nanostructures achieve the superior targeting ability, biocompatibility, and enhanced osteogenesis, providing a constructive design for tissue repair.


Subject(s)
Osteogenesis , Zeolites , Cell Differentiation , Cell Membrane , Stem Cells , Zeolites/chemistry
4.
Small ; 18(13): e2107236, 2022 04.
Article in English | MEDLINE | ID: mdl-35166031

ABSTRACT

Neural stem cells (NSCs) therapy is promising for treating neurodegenerative disorders and neural injuries. However, the limited in vitro expansion, spontaneous differentiation, and decrease in stemness obstruct the acquisition of high quantities of NSCs, restricting the clinical application of cell-based therapies and tissue engineering. This article reports a facile method of promoting NSCs expansion and maintaining stemness using wireless electrical stimulation triggered by piezoelectric nanomaterials. A nanofibrous membrane of poly L-lactic acid (PLLA) is prepared by electrostatic spinning, and the favorable piezoelectric property of PLLA facilitates the freeing of electrons after transformation. These self-powered electric signals generated by PLLA significantly enhance NSCs proliferation. Further, an undifferentiated cellular state is maintained in the NSCs cultured on the surfaces of PLLA nanofibers exposed to ultrasonic vibration. In addition, the neural differentiation potencies and functions of NSCs expanded by piezoelectric-driven localized electricity are not attenuated. Moreover, cell stemness can be maintained by wireless electric stimulation. Taken together, the electronic signals mediated by PLLA nanofibers facilitate NSCs proliferation. This efficient and simple strategy can maintain the stemness of NSCs during proliferation, which is essential for their clinical application, and opens up opportunities for the mass production of NSCs for use in cell therapy.


Subject(s)
Nanofibers , Neural Stem Cells , Cell Differentiation , Cell Proliferation , Lactic Acid , Polyesters , Tissue Engineering , Tissue Scaffolds
5.
Chem Soc Rev ; 50(22): 12828-12872, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34661592

ABSTRACT

One of the major issues in tissue engineering is regulation of stem cell differentiation toward specific lineages. Unlike biological and chemical signals, physical signals with adjustable properties can be applied to stem cells in a timely and localized manner, thus making them a hot topic for research in the fields of biomaterials, tissue engineering, and cell biology. According to the signals sensed by cells, physical signals used for regulating stem cell fate can be classified into six categories: mechanical, light, thermal, electrical, acoustic, and magnetic. In most cases, external macroscopic physical fields cannot be used to modulate stem cell fate, as only the localized physical signals accepted by the surface receptors can regulate stem cell differentiation via nanoscale fibrin polysaccharide fibers. However, surface receptors related to certain kinds of physical signals are still unknown. Recently, significant progress has been made in the development of functional materials for energy conversion. Consequently, localized physical fields can be produced by absorbing energy from an external physical field and subsequently releasing another type of localized energy through functional nanostructures. Based on the above concepts, we propose a methodology that can be utilized for stem cell engineering and for the regulation of stem cell fate via nanostructure-mediated physical signals. In this review, the combined effect of various approaches and mechanisms of physical signals provides a perspective on stem cell fate promotion by nanostructure-mediated physical signals. We expect that this review will aid the development of remote-controlled and wireless platforms to physically guide stem cell differentiation both in vitro and in vivo, using optimized stimulation parameters and mechanistic investigations while driving the progress of research in the fields of materials science, cell biology, and clinical research.


Subject(s)
Nanostructures , Stem Cells , Biocompatible Materials , Cell Differentiation , Tissue Engineering
6.
Int J Mol Sci ; 24(1)2022 Dec 28.
Article in English | MEDLINE | ID: mdl-36613973

ABSTRACT

Traditional bone defect treatments are limited by an insufficient supply of autologous bone, the immune rejection of allogeneic bone grafts, and high medical costs. To address this medical need, bone tissue engineering has emerged as a promising option. Among the existing tissue engineering materials, the use of electroactive scaffolds has become a common strategy in bone repair. However, single-function electroactive scaffolds are not sufficient for scientific research or clinical application. On the other hand, multifunctional electroactive scaffolds are often complicated and expensive to prepare. Therefore, we propose a new tissue engineering strategy that optimizes the electrical properties and biocompatibility of carbon-based materials. Here, a hydroxyapatite/carbon nanofiber (HAp/CNF) scaffold with optimal electrical activity was prepared by electrospinning HAp nanoparticle-incorporated polyvinylidene fluoride (PVDF) and then carbonizing the fibers. Biochemical assessments of the markers of osteogenesis in human adipose-derived stem cells (h-ADSCs) cultured on HAp/CNF scaffolds demonstrate that the material promoted the osteogenic differentiation of h-ADSCs in the absence of an osteogenic factor. The results of this study show that electroactive carbon materials with a fibrous structure can promote the osteogenic differentiation of h-ADSCs, providing a new strategy for the preparation and application of carbon-based materials in bone tissue engineering.


Subject(s)
Mesenchymal Stem Cells , Nanofibers , Humans , Osteogenesis , Tissue Scaffolds/chemistry , Durapatite/chemistry , Nanofibers/chemistry , Cells, Cultured , Tissue Engineering/methods , Cell Differentiation
7.
Molecules ; 27(6)2022 Mar 19.
Article in English | MEDLINE | ID: mdl-35335352

ABSTRACT

Oxidized low-density lipoprotein (oxLDL)-induced endothelium injury promotes the development of atherosclerosis. It has been reported that homoplantaginin, a flavonoid glycoside from the traditional Chinese medicine Salvia plebeia R. Br., protected vascular endothelial cells by inhibiting inflammation. However, it is undetermined whether homoplantaginin affects atherosclerosis. In this study, we evaluated the effect of homoplantaginin and its derivative dihydrohomoplantagin on oxLDL-induced endothelial cell injury and atherosclerosis in apoE-/- mice. Our results showedthat both dihydrohomoplantagin and homoplantaginin inhibited apoptosis and the increased level of ICAM-1 and VCAM-1 in oxLDL-stimulated HUVECs and the plaque endothelium of apoE-/- mice. Additionally, both of them restricted atherosclerosis development of apoE-/- mice. Mechanistic studies showed that oxLDL-induced the increase in ROS production, phosphorylation of ERK and nuclear translocation of NF-κB in HUVECs was significantly inhibited by the compounds. Meanwhile, these two compounds promoted Nrf2 nuclear translocation and increased the anti-oxidation downstream HO-1 protein level in HUVECs and plaque endothelium. Notably, knockdown of Nrf2 by siRNA abolished the cell protective effects of compounds and antagonized the inhibition effects of them on ROS production and NF-κB activation in oxLDL-stimulated HUVECs. Collectively, dihydrohomoplantagin and homoplantaginin protected VECs by activating Nrf2 and thus inhibited atherosclerosis in apoE-/- mice.


Subject(s)
Atherosclerosis , Salvia , Animals , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Endothelial Cells , Endothelium/metabolism , Flavonoids/pharmacology , Glucosides , Glycosides/metabolism , Glycosides/pharmacology , Lipoproteins, LDL/metabolism , Mice , NF-E2-Related Factor 2/metabolism , Salvia/metabolism , Signal Transduction
8.
World J Surg Oncol ; 19(1): 121, 2021 Apr 17.
Article in English | MEDLINE | ID: mdl-33865414

ABSTRACT

BACKGROUND: It has been reported that long-chain non-coding RNA (lncRNA) zinc finger E-box binding homeobox 1 antisense 1 (ZEB1-AS1) is an oncogene in various cancers, including hepatocellular carcinoma (HCC). We investigated the role and mechanism of ZEB1-AS1 as a competitive endogenous RNA (ceRNA) combined with miR-23c in HCC cell proliferation and invasion. METHODS: QRT-PCR was used to detect ZEB1-AS1 and miR-23c expressions in HCC tissues and cells. The dual luciferase reporter assay detected the targeted regulation of miR-23c and ZEB1-AS1. We also performed the correlation analysis of their expression in HCC tissues by the Spearman's correlation analysis. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to detect the proliferation of hepatoma cells. Cell invasion was assessed by the Transwell assay. RESULTS: QRT-PCR results indicated ZEB1-AS1 was upregulated and miR-23c was downregulated in HCC tissues and cell lines. ZEB1-AS1 knockdown hampered the proliferation and invasion of HCC cells. Dual luciferase reporter assay showed that miR-23c is a target of ZEB1-AS1, and ZEB1-AS1 was significantly negatively correlated with the miR-23c expression in HCC tissues. The results of MTT and Transwell assay showed that miR-23c inhibition restored the inhibitory effect of ZEB1-AS1 knockdown on HCC cells proliferation and invasion. CONCLUSIONS: As a ceRNA, lncRNA ZEB1-AS1 may play a vital role in inhibiting HCC progression through miR-23c, which will provide new clues and theoretical basis for the HCC diagnosis and treatment.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , RNA, Long Noncoding/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Apoptosis/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/pathology , MicroRNAs/genetics , Prognosis
9.
Nanotechnology ; 31(14): 145101, 2020 Apr 03.
Article in English | MEDLINE | ID: mdl-31846954

ABSTRACT

Recently, the wide application of upconversion nanoparticles (UCNPs) in the field of bioimaging has raised the requirement of biocompatibility. Current cytocompatibility studies on UCNPs mainly focus on cancer cells; however, their potential effects on normal cells are rarely addressed. Herein, the cellular effects of a trace amount of ligand-free NaYF4:Yb/Er nanocrystals on the differentiation of rat bone mesenchymal stem cells (rBMSCs) were investigated. First, due to their excellent upconversion fluorescent properties, the cellular uptake of ligand-free NaYF4:Yb/Er nanocrystals was confirmed by confocal laser scanning microscopy, and a homogeneous cytoplasmic distribution was imaged. Second, the viability of the rBMSCs cultured with a series of concentrations of nanoparticles (0, 30, 300, and 3000 ng ml-1) was evaluated, and a dose threshold was determined. Third, the effects of ligand-free NaYF4:Yb/Er nanocrystals on the osteogenesis of the rBMSCs were intensively characterized. The alkaline phosphatase activity assay, quantitative real time polymerase chain reaction for related osteogenic genes, and immunofluorescence staining of specific biomarkers and mineral deposits demonstrated that the ligand-free NaYF4:Yb/Er nanocrystals at a proper concentration can enhance osteogenic differentiation. Finally, intracytoplasmic lipid detection showed that the adipogenic differentiation of rBMSCs might be inhibited in the presence of ligand-free NaYF4:Yb/Er nanocrystals. Meanwhile, these results showed that the effects of ligand-free NaYF4:Yb/Er nanocrystals on rBMSCs were concentration-dependent and reciprocal between osteogenic and adipogenic differentiation. This work provides new insights into the exploring the biocompatibility of UCNPs and will benefit the research community engaged in nanotechnology and biomedicine.


Subject(s)
Adipogenesis/drug effects , Erbium/chemistry , Fluorides/pharmacology , Mesenchymal Stem Cells/cytology , Osteogenesis/drug effects , Ytterbium/chemistry , Yttrium/pharmacology , Animals , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Fluorides/chemistry , HeLa Cells , Humans , Ligands , Mesenchymal Stem Cells/drug effects , Microscopy, Confocal , Nanoparticles , Rats , Staining and Labeling , Yttrium/chemistry
10.
Am J Physiol Cell Physiol ; 316(1): C70-C80, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30404560

ABSTRACT

Myasthenia gravis (MG) is an autoimmune neuromuscular disorder, affecting the quality of life of millions of people worldwide. The present study aims to determine the relationship between micro-RNA-143 (miR-143) and C-X-C motif chemokine 13 (CXCL13) and whether it influences the pathogenesis of myasthenia gravis (MG). Thymus specimens were resected from patients with thymic hyperplasia combined with MG and then infused into normal mouse cavities to establish MG mouse models. Immunohistochemistry, reverse transcription-quantitative PCR, in situ hybridization detection, and Western blot analysis were employed to identify the expression of miR-143 and CXCL13 in MG and normal mice. The obtained thymocytes were cultured in vitro and transfected with a series of miR-143 mimic, miR-143 inhibitor, overexpression of CXCL13, or siRNA against CXCL13. MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] and flow cytometry assays were employed to assess cell viability, cycle entry, and apoptosis of the thymocytes. Dual-luciferase reporter assay provided verification, confirming that CXCL13 was the target gene of miR-143. Low miR-143 expression in the thymus tissues of the MG mice was detected, which presented with a reciprocal relationship with the expression rate of CLCX13. Observations in relation to the interactions between miR-143 mimic or siRNA-CXCL13 exposure showed reduced cell viability, with a greater number of cells arrested at the G0/G1 phase and a greater rate of induced apoptosis. Furthermore, overexpression of CXCL13 rescued miR-143 mimic-induced apoptosis. The findings have identified the potential role of miR-143 as a MG development mediator by targeting CXCL13. The key results obtained provide a promising experimental basis for targeted intervention treatment with miR-143.


Subject(s)
Cell Proliferation/physiology , Chemokine CXCL13/biosynthesis , Disease Models, Animal , MicroRNAs/biosynthesis , Myasthenia Gravis/metabolism , Thymocytes/metabolism , Adolescent , Adult , Animals , Apoptosis/physiology , Cells, Cultured , Chemokine CXCL13/antagonists & inhibitors , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Myasthenia Gravis/pathology , Thymocytes/pathology , Young Adult
11.
J Cell Physiol ; 234(5): 5926-5939, 2019 05.
Article in English | MEDLINE | ID: mdl-30585631

ABSTRACT

Autophagy is a vital negative factor regulating cellular senescence. Purple sweet potato color (PSPC), one type of flavonoid, has been demonstrated to suppress endothelial senescence and restore endothelial function in diabetic mice by inhibiting the nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain containing protein 3 (NLRP3) inflammasome. However, the roles of autophagy in the inflammatory response during endothelial senescence are unknown. Here, we found that PSPC augmented autophagy to restrict high-glucose-induced premature endothelial senescence. In addition, PSPC administration impaired endothelium aging in diabetic mice by increasing autophagy. Inhibition of autophagy accelerated endothelial senescence, while enhancement of autophagy delayed senescence. Moreover, deactivation of the NLRP3 inflammasome triggered by PSPC was autophagy-dependent. Autophagy receptor microtubule-associated protein 1 light chain 3 and p62 interacted with the inflammasome component NLRP3, suggesting that autophagosomes target the NLRP3 inflammasome and deliver it to the lysosome for degradation. Altogether, PSPC amplified cellular autophagy, subsequently attenuated NLRP3 inflammasome activity and finally delayed endothelial senescence to ameliorate cardiovascular complication. These results suggest a potential therapeutic target in senescence-related cardiovascular diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Autophagy/drug effects , Cellular Senescence/drug effects , Flavonoids/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Ipomoea batatas , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pigments, Biological/pharmacology , Animals , Anti-Inflammatory Agents/isolation & purification , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy-Related Proteins/metabolism , Cells, Cultured , Cytokines/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat , Disease Models, Animal , Flavonoids/isolation & purification , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Ipomoea batatas/chemistry , Lysosomes/metabolism , Male , Mice, Inbred ICR , Pigments, Biological/isolation & purification , Protein Denaturation , Protein Transport , Signal Transduction
12.
J Cell Physiol ; 234(6): 9033-9044, 2019 06.
Article in English | MEDLINE | ID: mdl-30362546

ABSTRACT

Development of effective therapeutic drugs for Parkinson's disease (PD) is of great importance. Aberrant microRNA (miRNA) expression has been identified in postmortem human PD brain samples, in vitro and in vivo PD models. However, the role of miR-342-3p in PD has been understudied. The study explores the effects of miR-342-3p on expression of glutamate (Glu) transporter, and dopaminergic neuron apoptosis and proliferation by targeting p21-activated kinase 1 (PAK1) through the Wnt signaling pathway in PD mice. After establishment of PD mouse models, gain- or loss-of-function assay was performed to explore the functional role of miR-342-3p in PD. Number of apoptotic neurons and Glu concentration was then determined. Subsequently, PC12 cells were treated with miR-342-3p mimic, miR-342-3p inhibitor, dickkopf-1 (DKK1), and miR-342-3p inhibitor + DKK1. The expression of miR-342-3p, PAK1, the Wnt signaling pathway-related and apoptosis-related genes, Glutamate transporter subtype 1 (GLT-1), l-glutamate/ l-aspartate transporter (GLAST), tyrosine hydroxylase (TH) was measured. Also, cell viability and apoptosis were evaluated. PD mice exhibited increased miR-342-3p, while decreased expression of PAK1, GLT-1, GLAST, TH, and the Wnt signaling pathway-related and antiapoptosis genes. miR-342-3p downregulation could promote expression of PAK1, the Wnt signaling pathway-related and antiapoptosis genes. GLT-1, GLAST, and TH as well as cell viability, but reduce cell apoptosis rate. The results indicated that suppression of miR-342-3p improves expression of Glu transporter and promotes dopaminergic neuron proliferation while suppressing apoptosis through the Wnt signaling pathway by targeting PAK1 in mice with PD.


Subject(s)
Apoptosis , Brain/enzymology , Dopaminergic Neurons/enzymology , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/metabolism , MicroRNAs/metabolism , Parkinson Disease/enzymology , Wnt Signaling Pathway , p21-Activated Kinases/metabolism , Animals , Brain/pathology , Cell Proliferation , Disease Models, Animal , Dopaminergic Neurons/pathology , Down-Regulation , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 2/genetics , Gene Expression Regulation, Enzymologic , Male , Mice, Inbred C57BL , MicroRNAs/genetics , PC12 Cells , Parkinson Disease/genetics , Parkinson Disease/pathology , Rats , p21-Activated Kinases/genetics
13.
J Cell Biochem ; 120(1): 861-871, 2019 01.
Article in English | MEDLINE | ID: mdl-30125994

ABSTRACT

Ectodermal-neural cortex 1 (ENC1) belongs to a member of the kelch family of genes. It is an actin-binding protein and plays a pivotal role in neuronal and adipocyte differentiation. Here, we found that lower expression of ENC1 in the ovarian cancer patients was associated with favorable prognosis. In addition, ENC1 was heterogeneously expressed in various ovarian cancer cells. The messenger RNA and protein expression levels of ENC1 in HO-8910PM and NIH:OVCAR-3 cells were obviously higher than that in the other types of ovarian cancer cells. Knockdown of ENC1 in HO-8910PM or NIH:OVCAR-3 cells could significantly increase the reactive oxygen species levels, resulting in inhibition of in vitro proliferation, migration, and invasion. Our findings suggest that decreasing expression of ENC1 may be a new approach that can be used for ovarian cancer treatment.


Subject(s)
Gene Expression Regulation, Neoplastic , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cohort Studies , Female , Gene Knockdown Techniques , Humans , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Staging , Ovarian Neoplasms/metabolism , Prognosis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Transfection
14.
Mol Med ; 25(1): 29, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31215394

ABSTRACT

BACKGROUND: Innate immune dysfunction contributes to the development and progression of nonalcoholic fatty liver disease (NAFLD), however, its pathogenesis is still incompletely understood. Identifying the key innate immune component responsible for the pathogenesis of NAFLD and clarifying the underlying mechanisms may provide therapeutic targets for NAFLD. Recently, F-box- and WD repeat domain-containing 7 (FBXW7) exhibits a regulatory role in hepatic glucose and lipid metabolism. This study aims to investigate whether FBXW7 controls high-mobility group box 1 protein (HMGB1)-mediated innate immune signaling to improve NAFLD and the mechanism underlying this action. METHODS: Mice were fed a high-fat diet (HFD) for 12 or 20 weeks to establish NAFLD model. Hepatic overexpression or knockdown of FBXW7 was induced by tail-vein injection of recombinant adenovirus. Some Ad-FBXW7-injected mice fed a HFD were injected intraperitoneally with recombinant mouse HMGB1 to confirm the protective role of FBXW7 in NAFLD via inhibition of HMGB1. RESULTS: FBXW7 improves NAFLD and related metabolic parameters without remarkable influence of body weight and food intake. Moreover, FBXW7 markedly ameliorated hepatic inflammation and insulin resistance in the HFD-fed mice. Furthermore, FBXW7 dramatically attenuated the expression and release of HMGB1 in the livers of HFD-fed mice, which is associated with inhibition of protein kinase R (PKR) signaling. Thereby, FBXW7 restrains Toll-like receptor 4 (TLR4) and receptor for advanced glycation end products (RAGE) signaling in HFD-fed mouse livers. In addition, exogenous HMGB1 treatment abolished FBXW7-mediated inhibition of hepatic inflammation and insulin resistance in HFD-fed mouse livers. CONCLUSIONS: Our results demonstrate a protective role of FBXW7 in NAFLD by abating HMGB1-mediated innate immune signaling to suppress inflammation and consequent insulin resistance, suggesting that FBXW7 is a potential target for therapeutic intervention in NAFLD development.


Subject(s)
F-Box-WD Repeat-Containing Protein 7/metabolism , HMGB1 Protein/metabolism , Liver/metabolism , Mice, Inbred C57BL/physiology , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Blotting, Western , F-Box-WD Repeat-Containing Protein 7/genetics , Fluorescent Antibody Technique , Glucose Tolerance Test , HMGB1 Protein/genetics , Immunity, Innate/genetics , Immunohistochemistry , Insulin Resistance/genetics , Insulin Resistance/physiology , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Male , Mice , Mice, Inbred C57BL/genetics , Non-alcoholic Fatty Liver Disease/genetics , Signal Transduction/genetics , Signal Transduction/physiology
15.
IUBMB Life ; 71(1): 81-92, 2019 01.
Article in English | MEDLINE | ID: mdl-30296359

ABSTRACT

Glioma is known to be the most prevalent primary brain tumor. In recent years, there has been evidence indicating myeloid cell leukemia-1 (MCL1) plays a role in brain glioblastoma. Therefore, the present study was conducted with aims of exploring the ability of MCL1 silencing to influence glioma cell senescence and apoptosis through the mediation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway. Glioma and tumor-adjacent tissues were collected in order to detect the presence of higher levels of MCL1 protein expression. Next, the mRNA and protein expression of MCL1, PI3K, Akt, B cell lymphoma 2 (Bcl2), Bcl2-associated X (Bax), B lymphoma Mo-MLV insertion region 1 homolog (Bmi-1), and phosphatase and tensin homolog (PTEN) were determined. Cell counting kit-8 assay was applied to detect cell proliferation, ß-galactosidase staining for cell senescence, and flow cytometry for cell cycle entry and apoptosis. Initially, the results revealed higher positive expression rate of MCL1 protein, increased mRNA and protein expression of MCL1, PI3K, Akt, Bmi-1, and Bcl-2 and decreased that of Bax and PTEN in human glioma tissues. The silencing of MCL1 resulted in a decrease in mRNA and protein expression of PI3K, Akt, Bmi-1, and Bcl-2 and an increase in Bax and PTEN expressions in glioma cells. Moreover, silencing of MCL1 also inhibited cell proliferation and cell cycle entry in glioma cells, and promoted glioma cell senescence and apoptosis. In conclusion, the aforementioned results collectively suggested that the silencing of MCL1 promotes senescence and apoptosis in glioma cells through inhibiting the PI3K/Akt signaling pathway. Thus, decreasing the expression of MCL1 might have therapeutic functions in glioma. © 2018 IUBMB Life, 71(1):81-92, 2019.


Subject(s)
Cell Proliferation/genetics , Cellular Senescence/genetics , Glioma/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Adolescent , Adult , Apoptosis/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing , Glioma/pathology , Humans , Male , Middle Aged , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction/genetics , Young Adult , bcl-2-Associated X Protein/genetics
16.
Zhongguo Dang Dai Er Ke Za Zhi ; 21(10): 972-976, 2019 Oct.
Article in Zh | MEDLINE | ID: mdl-31642429

ABSTRACT

OBJECTIVE: To study the types and characteristics of TUBB1 mutation in children with congenital hypothyroidism (CH) and thyroid dysgenesis (TD) in Shandong, China. METHODS: Mutations of the whole coding region of the TUBB1 gene were analyzed for 289 children with CH and TD in Shandong. Whole-genome DNA was extracted from peripheral blood leukocytes. PCR multiplication was performed for the whole coding region of the TUBB1 gene. Sanger sequencing was performed for the PCR products, and a biological information analysis was performed. RESULTS: Among the 289 children with CH and TD, 4 (1.4%) were found to have a c.952C>T(p.R318W) heterozygous mutation in the TUBB1 gene, resulting in the change of tryptophan into arginine at codon 318 of TUBB1 protein. This mutation was evaluated as "potentially pathogenic" based on the classification criteria and guidelines for genetic variation by American College of Medical Genetics and Genomics. CONCLUSIONS: A novel mutation is detected in the exon of the TUBB1 gene in children with CH and TD in Shandong, suggesting that the TUBB1 gene may be a candidate pathogenic gene for CH children with TD.


Subject(s)
Congenital Hypothyroidism , Thyroid Dysgenesis , Tubulin/genetics , Child , China , Congenital Hypothyroidism/genetics , DNA Mutational Analysis , Humans , Mutation , Thyroid Dysgenesis/genetics
17.
J Cell Mol Med ; 22(6): 3167-3182, 2018 06.
Article in English | MEDLINE | ID: mdl-29536658

ABSTRACT

Hypoxia-ischaemia (HI) remains a major cause of foetal brain damage presented a scarcity of effective therapeutic approaches. Dexmedetomidine (DEX) and microRNA-140-5p (miR-140-5p) have been highlighted due to its potentially significant role in the treatment of cerebral ischaemia. This study was to investigate the role by which miR-140-5p provides cerebral protection using DEX to treat hypoxic-ischaemic brain damage (HIBD) in neonatal rats via the Wnt/ß-catenin signalling pathway. The HIBD rat models were established and allocated into various groups with different treatment plans, and eight SD rats into sham group. The learning and memory ability of the rats was assessed. Apoptosis and pathological changes in the hippocampus CA1 region and expressions of the related genes of the Wnt/ß-catenin signalling pathway as well as the genes responsible of apoptosis were detected. Compared with the sham group, the parameters of weight, length growth, weight ratio between hemispheres, the rate of reaching standard, as well as Bcl-2 expressions, were all increased. Furthermore, observations of increased levels of cerebral infarction volume, total mortality rate, response times, total response duration, expressions of Wnt1, ß-catenin, TCF-4, E-cadherin, apoptosis rate of neurons, and Bax expression were elevated. Following DEX treatment, the symptoms exhibited by HIBD rats were ameliorated. miR-140-5p and si-Wnt1 were noted to attenuate the progression of HIBD. Our study demonstrates that miR-140-5p promotes the cerebral protective effects of DEX against HIBD in neonatal rats by targeting the Wnt1 gene through via the negative regulation of the Wnt/ß-catenin signalling pathway.


Subject(s)
Dexmedetomidine/administration & dosage , Hypoxia, Brain/drug therapy , Hypoxia-Ischemia, Brain/drug therapy , MicroRNAs/genetics , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Proliferation/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Gene Expression Regulation/drug effects , Humans , Hypoxia, Brain/genetics , Hypoxia, Brain/pathology , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/pathology , Neurons/drug effects , Neurons/pathology , Rats , Wnt Signaling Pathway , Wnt1 Protein/genetics , beta Catenin/genetics
18.
J Cell Physiol ; 233(12): 9488-9502, 2018 12.
Article in English | MEDLINE | ID: mdl-29995978

ABSTRACT

Recent studies have proposed that microRNAs (miR) function as novel diagnostic and prognostic biomarkers and therapeutic targets in Alzheimer's disease (AD), a common disease among the elderly. In the current study, we aim to explore the effect of miR-186 on oxidative stress injury of neuron in rat models of AD with the involvement of the interleukin-2 (IL2) and the Janus kinase/signal transducers and activators of transcription (JAK-STAT) pathways. AD rat models were established, and dual-luciferase reporter assay and online software were used to confirm the targeting relationship between miR-186 and IL2. Immunohistochemistry was used evaluating the positive rate of IL2. Afterward, to define the role of miR-186 in AD, miR-186, IL2, and JAK-STAT related protein (JAK2, STAT3) expressions were quantified. Cell proliferation was measured by 3-(4,5-dimethylthiazol-2-yl)2,5-diphenyl tetrazolium bromide, and cell apoptosis was detected by flow cytometry. We observed downregulated miR-186 and IL2 and upregulated JAK-STAT signaling pathway related genes in AD. The overexpression of miR-186 was shown to significantly promote cell proliferation while suppressing cell apoptosis along with the expression of the IL2 and JAK-STAT signaling pathway related protein. Collectively, the key findings obtained from the current study define the potential role of miR-186 as an inhibitor of AD development by downregulation of IL2 through suppression of the JAK-STAT signaling pathway.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Interleukin-2/metabolism , Janus Kinases/metabolism , MicroRNAs/metabolism , Neurons/pathology , Oxidative Stress , STAT3 Transcription Factor/metabolism , Alzheimer Disease/physiopathology , Animals , Apoptosis , Base Sequence , Caspase 3/metabolism , Disease Models, Animal , Down-Regulation , Epidermal Growth Factor/metabolism , Glutathione Peroxidase/metabolism , Growth Hormone/metabolism , Hippocampus/pathology , Interferon-gamma/metabolism , Interleukin-2/genetics , L-Lactate Dehydrogenase/metabolism , Male , Malondialdehyde/metabolism , Memory Disorders/genetics , Memory Disorders/pathology , MicroRNAs/genetics , Neurons/metabolism , Platelet-Derived Growth Factor/metabolism , Rats, Sprague-Dawley , Reaction Time , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , bcl-2-Associated X Protein/metabolism
19.
J Cell Physiol ; 233(9): 7022-7034, 2018 09.
Article in English | MEDLINE | ID: mdl-29380367

ABSTRACT

Epilepsy is a group of neurological disorders characterized by epileptic seizures. In this study, we aim to explore the role of microRNA-421 (miR-421) in hippocampal neurons of epilepsy mice via the TLR/MYD88 pathway. Forty mice were randomly served as the normal and model (established as epilepsy model) groups. Hippocampal neurons were assigned into seven groups with different transfections. The RT-qPCR and western blotting were conducted to examine the expression of miR-421 TLR2, TLR4, MYD88, Bax, Bcl-2, p53, Beclin-1, and LC3II/LC3I. Cell proliferation and apoptosis were detected by MTT and flow cytometry.MYD88 is a target gene of miR-421. Model mice showed elevated expression of TLR2, TLR4, MYD88, Bax, p53, Beclin-1, and LC3II/LC3I but reduced expression of miR-421 and Bcl-2. In vitro experiments reveals that overexpression of miR-421 inhibited the TLR/MYD88 pathway. Besides, overexpressed miR-421 declined cell apoptosis but increased cell proliferation. It reveals that miR-421 targeting MYD88 could inhibit the apoptosis and autophagy of hippocampal neurons in epilepsy mice by down-regulating the TLR/MYD88 pathway.


Subject(s)
Apoptosis , Autophagy , Epilepsy/genetics , Hippocampus/pathology , MicroRNAs/metabolism , Myeloid Differentiation Factor 88/metabolism , Neurons/pathology , Toll-Like Receptors/metabolism , Animals , Apoptosis/genetics , Autophagy/genetics , Base Sequence , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/ultrastructure , Cell Cycle Checkpoints , Cell Proliferation/genetics , Disease Models, Animal , Epilepsy/pathology , Male , Mice , MicroRNAs/genetics , Neurons/metabolism , RNA, Small Interfering/metabolism , S Phase , Signal Transduction
20.
J Cell Physiol ; 233(8): 5895-5907, 2018 08.
Article in English | MEDLINE | ID: mdl-29227541

ABSTRACT

This study investigates the protective effects of miR-431 against cerebral ischemia-reperfusion injury through the Rho/Rho-kinase signaling pathway. SD rats were randomly classified into normal, sham, and model (middle cerebral artery occluded) groups. Rho expression and cerebral infarction were visualized by immunohischemistry and TTC staining, respectively. qRT-PCR and western blotting were used to measure mRNA and protein expression of miR-431 and Rho/Rho-kinase signaling pathway-related genes. Hippocampal neurons were extracted and assigned into normal, blank, negative control (NC), miR-431 mimics, miR-431 inhibitors, siRNA-Rho, and miR-431 inhibitors + siRNA-Rho groups. Proliferation and apoptosis were detected by MTT and flow cytometry, respectively. Compared with the normal group, the model group showed elevated Rho expression, area of cerebral infarction, and expressions of Rho/Rho-kinase related genes but reduced miR-431 expression. Compared with the blank group, expression of Rho, Rho-kinase α, and Rho-kinase ß decreased and miR-431 expression increased in the miR-431 mimics and siRNA-Rho groups, and the tendency reversed in the miR-431 inhibitors group. Enhanced proliferation and inhibited apoptosis were exhibited in the miR-431 mimics and siRNA-Rho groups while results in the miR-431 inhibitors group reversed. Findings obtained from this study indicated that miR-431 confers protection against cerebral ischemia-reperfusion injury through negatively regulating the Rho/Rho-kinase signaling pathway.


Subject(s)
Cerebral Infarction/prevention & control , Hippocampus/pathology , MicroRNAs/genetics , Reperfusion Injury/prevention & control , rho-Associated Kinases/metabolism , Animals , Apoptosis/genetics , Cell Proliferation/genetics , Cells, Cultured , Cerebral Infarction/pathology , Disease Models, Animal , Hippocampus/cytology , Male , RNA Interference , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/genetics , rho-Associated Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL