Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Mol Carcinog ; 63(8): 1572-1587, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38780170

ABSTRACT

Clear cell renal cell carcinoma (ccRCC), the most common subtype of renal cell carcinoma, often leads to a poor prognosis due to metastasis. The investigation of N6-methyladenosine (m6A) methylation, a crucial RNA modification, and its role in ccRCC, particularly through the m6A reader insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2), revealed significant insights. We found that IGF2BP2 was notably downregulated in ccRCC, which correlated with tumor aggressiveness and poor prognosis. Thus, IGFBP2 has emerged as an independent prognostic factor of ccRCC. Moreover, a strong positive correlation was observed between the expression of IGF2BP2 and Netrin-4. Netrin-4 was also downregulated in ccRCC, and its lower levels were associated with increased malignancy and poor prognosis. Overexpression of IGF2BP2 and Netrin-4 suppressed the invasion and migration of ccRCC cells, while Netrin-4 knockdown reversed these effects in ccRCC cell lines. RNA immunoprecipitation (RIP)-quantitative polymerase chain reaction validated the robust enrichment of Netrin-4 mRNA in anti-IGF2BP2 antibody immunoprecipitates. MeRlP showed significantly increased Netrin4 m6A levels after lGF2BP2 overexpression. Moreover, we found that IGF2BP2 recognized and bound to the m6A site within the coding sequence of Netrin-4, enhancing its mRNA stability. Collectively, these results showed that IGF2BP2 plays a suppressive role in the invasion and migration of ccRCC cells by targeting Netrin-4 in an m6A-dependent manner. These findings underscore the potential of IGF2BP2/Netrin-4 as a promising prognostic biomarker and therapeutic target in patients with ccRCC metastasis.


Subject(s)
Carcinoma, Renal Cell , Cell Movement , Gene Expression Regulation, Neoplastic , Kidney Neoplasms , Neoplasm Invasiveness , Netrins , RNA-Binding Proteins , Humans , Netrins/genetics , Netrins/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/metabolism , Kidney Neoplasms/genetics , Prognosis , Cell Line, Tumor , Male , Adenosine/analogs & derivatives , Adenosine/metabolism , Female , Cell Proliferation , Middle Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism
2.
J Neurochem ; 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38115597

ABSTRACT

Diosgenin, a natural steroid saponin, holds promise as a multitarget therapeutic for various diseases, including neurodegenerative conditions. Its efficacy in slowing Alzheimer's disease, Parkinson's disease, multiple sclerosis, and stroke progression has been demonstrated. However, the role of diosgenin in anti-epilepsy and its potential connection to the modulation of the intestinal microbiota remain poorly understood. In this study, exogenous diosgenin significantly mitigated pentylenetetrazole (PTZ)-induced seizures, learning and memory deficits, and hippocampal neuronal injury. 16S ribosomal RNA (16S rRNA) sequencing revealed a reversal in the decrease of Bacteroides and Parabacteroides genera in the PTZ-induced mouse epileptic model following diosgenin treatment. Fecal microbiota transplantation (FMT) experiments illustrated the involvement of diosgenin in modulating gut microbiota and providing neuroprotection against epilepsy. Our results further indicated the repression of enteric glial cells (EGCs) activation and the TLR4-MyD88 pathway, coupled with reduced production of inflammatory cytokines in the colonic lumen, and improved intestinal barrier function in epilepsy mice treated with diosgenin or FMT. This study suggests that diosgenin plays a role in modifying gut microbiota, contributing to the alleviation of intestinal inflammation and neuroinflammation, ultimately inhibiting epilepsy progression in a PTZ-induced mouse model. Diosgenin emerges as a potential therapeutic option for managing epilepsy and its associated comorbidities.

3.
Cell Mol Neurobiol ; 43(5): 1663-1683, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36350538

ABSTRACT

Netrin-4, a member of the Netrins family, is an important secreted protein that plays a role in axonal outgrowth and migration orientation. It was initially described that Netrin-4 had a high correlation with the laminin ß-chain and promoted the growth of neurites in cultured olfactory bulb explants. Subsequently, it was discovered that Netrin-4 is involved in regulating various physiological processes, including angiogenesis, the occurrence and metastasis of various tumors, and the development of the kidney and alveoli. This paper reviews the current research on Netrin-4 since its discovery and provides a theoretical basis for further research on the biological characteristics of Netrin-4. Effects of Netrin-4. Netrin-4 regulates axon guidance, angiogenesis and the development of various tumors.


Subject(s)
Neoplasms , Receptors, Cell Surface , Humans , Receptors, Cell Surface/metabolism , Nerve Growth Factors/pharmacology , Nerve Growth Factors/metabolism , Axon Guidance , Tumor Suppressor Proteins/metabolism , Netrins , Axons/metabolism
4.
Am J Physiol Endocrinol Metab ; 323(5): E405-E417, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36103628

ABSTRACT

Polycystic ovary syndrome (PCOS) is an extremely common endocrine-metabolic disorder and the main cause of infertility in premenopausal women, thus targeted treatments are sorely needed. Accumulative evidence showed that exogenous supplementation of IL-22 in PCOS mice may be of significant positive effect on insulin resistance (IR), a root causative factor for this condition, but much remained unknown about its mechanism. According to our previous study, troxerutin, a common anticoagulant and thrombolytic agent in clinic, alleviated various PCOS-like phenotypes in dihydrotestosterone (DHT)-treated rat model with unclear mechanism. Here, glucose tolerance tests (GTTs), insulin tolerance tests (ITTs), and homeostatic model assessment of insulin resistance (HOMA-IR) analyses revealed that troxerutin treatment in DHT-treated rats also significantly improved insulin resistance and enhanced serum IL-22 levels, which thereby activated IL-22R1/Janus kinase 1 (JAK1)/signal transducer and activator of transcription-3 (STAT3) signaling pathway in pancreatic islet. This protective effect of troxerutin on insulin resistance improvement was blocked by an inhibitor of p-STAT3, S3I-201. Troxerutin administration to DHT rats decreased the relative abundance of Bifidobacterium and enhanced secondary bile acid profiles, which were positively correlated with serum IL-22 concentration. Conclusively, the present study reported that troxerutin is an endogenous enhancer of IL-22 and the effect of troxerutin on insulin resistance improvement was via IL-22R1/JAK1/STAT3 signaling activation in a DHT-induced PCOS rat model. These insights may be translated into a primary therapeutic agent for PCOS with insulin resistance and hyperandrogenism.NEW & NOTEWORTHY Troxerutin decreased the relative abundance of Bifidobacterium, along with enhancement of secondary bile acids/IL-22 system, which thereby activated its downstream IL-22R1/JAK1/STAT3 signaling pathway in pancreatic ß cells, subsequently attenuated insulin resistance (IR), hyperandrogenism and PCOS-like phenotypes in DHT-induced PCOS rat models. Troxerutin is an endogenous IL-22 enhancer and may be of therapeutic value for PCOS with insulin resistance.


Subject(s)
Hyperandrogenism , Insulin Resistance , Polycystic Ovary Syndrome , Animals , Female , Humans , Mice , Rats , Anticoagulants , Bile Acids and Salts/pharmacology , Dihydrotestosterone/pharmacology , Fibrinolytic Agents , Insulin/metabolism , Insulin Resistance/physiology , Janus Kinase 1/metabolism , Janus Kinase 1/pharmacology , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/drug therapy , Signal Transduction , STAT3 Transcription Factor/metabolism , Interleukin-22
5.
J Cell Physiol ; 235(4): 3939-3949, 2020 04.
Article in English | MEDLINE | ID: mdl-31595977

ABSTRACT

Ovarian cancer (OC) is a highly prevalent gynecologic malignancy and its mortality is extremely high. Therefore, the development of novel therapeutic approaches for OC is of great significance. In this study, LINC01342 was upregulated in OC tissue in the GSE38666 microarray and in tumor tissue samples collected in our center. The silencing of LINC01342 suppressed the proliferative and metastatic capacities of A2780 and HO8910 cells. Subcellular distribution assays showed that LINC01342 was mainly enriched in the cytoplasm. Subsequently, the downregulation of microRNA-30c-2-3p was proven to be the target of LINC01342. The silencing of microRNA-30c-2-3p enhanced the clonality and migratory capacity of OC cells. Moreover, the silencing of microRNA-30c-2-3p could reverse the inhibited migration and clonality in OC cells caused by LINC01342 knockdown. In addition, hypoxia-inducible factor 3 subunit α (HIF3A) was proven to be the target gene of microRNA-30c-2-3p, which was upregulated. HIF3A was negatively regulated by microRNA-30c-2-3p but positively regulated by LINC01342 in OC cells. An RNA binding protein immunoprecipitation assay showed that microRNA-30c-2-3p, LINC01342, and HIF3A could bind to argonaute RISC catalytic component 2. The overexpression of HIF3A reversed the inhibited migration and clonality in OC cells with LINC01342 knockdown. By analyzing the follow-up data from the enrolled OC patients, the LINC01342 and HIF3A levels were negatively correlated with prognosis, while the microRNA-30c-2-3p level was positively correlated with the same. In short, the upregulated LINC01342 in OC absorbs microRNA-30c-2-3p to release HIF3A. Thus, upregulated HIF3A expression accelerates the progression of OC.


Subject(s)
Apoptosis Regulatory Proteins/genetics , MicroRNAs/genetics , Ovarian Neoplasms/genetics , RNA, Long Noncoding/genetics , Repressor Proteins/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , Ovarian Neoplasms/pathology
6.
J Neuroinflammation ; 16(1): 183, 2019 Sep 27.
Article in English | MEDLINE | ID: mdl-31561751

ABSTRACT

BACKGROUND: Toll-like receptor 4 (TLR4) is well known for activating the innate immune system; however, it is also highly expressed in adaptive immune cells, such as CD4+ T-helper 17 (Th17) cells, which play a key role in multiple sclerosis (MS) pathology. However, the function and governing mechanism of TLR4 in Th17 remain unclear. METHODS: The changes of TLR4 in CD4+ T cells from MS patients and experimental autoimmune encephalomyelitis (EAE) mice were tested. TLR4-deficient (TLR4-/-) naïve T cells were induced in vitro and transferred into Rag1-/- mice to measure Th17 differentiation and EAE pathology. DNA sequence analyses combining with deletion fragments and mutation analyses, chromatin immunoprecipitation (ChIP), and electrophoretic mobility shift assay (EMSA) were used to explore the mechanism of TLR4 signaling pathway in regulating Th17 differentiation. RESULTS: The levels of TLR4 were increased in CD4+ Th17 cells both from MS patients and EAE mice, as well as during Th17 differentiation in vitro. TLR4-/- CD4+ naïve T cells inhibited their differentiation into Th17, and transfer of TLR4-/- CD4+ naïve T cells into Rag1-/- mice was defective in promoting EAE, characterized by less demyelination and Th17 infiltration in the spinal cord. TLR4 signal enhanced Th17 differentiation by activating RelA, downregulating the expression of miR-30a, a negative regulator of Th17 differentiation. Inhibition of RelA activity increased miR-30a level, but decreased Th17 differentiation rate. Furthermore, RelA directly regulated the expression of miR-30a via specific binding to a conserved element of miR-30a gene. CONCLUSIONS: TLR4-/- CD4+ naïve T cells are inadequate in differentiating to Th17 cells both in vitro and in vivo. TLR4-RelA-miR-30a signal pathway regulates Th17 differentiation via direct binding of RelA to the regulatory element of miR-30a gene. Our results indicate modulating TLR4-RelA-miR-30a signal in Th17 may be a therapeutic target for Th17-mediated neurodegeneration in neuroinflammatory diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Signal Transduction/physiology , Th17 Cells/immunology , Animals , Cell Differentiation/immunology , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/immunology , MicroRNAs/metabolism , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Transcription Factor RelA/immunology , Transcription Factor RelA/metabolism
7.
Int J Neurosci ; 129(9): 833-841, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30668212

ABSTRACT

Objectives: Parkinson's disease (PD) is an age-related neurodegenerative disease characterized by motor dysfunctions. Dopaminergic neuron loss, inflammation and oxidative stress responses play key roles in the pathogenisis of PD. Osthole (Ost), a natural coumarin derivative, isolated from various herbs such as Cnidium monnieri (L.), has anti-inflammatory, anti-apoptotic and anti-oxidative stress properties. However, whether it has effects on PD is unknown. Methods: In this study, mice were subjected to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) injection to induce PD symptoms, and treated with osthole. Stepping and cylinder tests were performed to determine their motor function. Immunohistochemical and immunofluorescence staining were performed to detect tyrosine hydroxylase (TH) and ionized calcium binding adaptor molecule 1 (Iba-1). The expression levels of inflammatory cytokines and oxidative stress factors were detected by qPCR and ELISA. Notch signaling pathway was investigated by western blot. Results: We found that injection of MPTP induced motor deficits in mice, enhanced the loss dopaminergic neurons and the activation of microglia, increased inflammatory and oxidative stress responses, and inhibited Notch signaling pathway. Osthole treatment suppressed theses MPTP-induced alterations. Conclusion: In conclusion, osthole attenuates PD symptoms by suppressing Notch signaling pathway.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Coumarins/therapeutic use , MPTP Poisoning/drug therapy , Receptors, Notch/antagonists & inhibitors , Signal Transduction/drug effects , Adjuvants, Immunologic/pharmacology , Animals , Coumarins/pharmacology , MPTP Poisoning/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , PC12 Cells , Random Allocation , Rats , Receptors, Notch/metabolism , Signal Transduction/physiology
8.
Eur J Neurosci ; 45(2): 249-259, 2017 01.
Article in English | MEDLINE | ID: mdl-27873367

ABSTRACT

Remyelination is limited in patients with multiple sclerosis (MS) due to the difficulties in recruiting proliferating oligodendrocyte precursors (OPCs), the inhibition of OPC differentiation and/or maturation, and/or failure in the generation of the myelin sheath. In vitro studies have revealed that miR-219 is necessary for OPC differentiation and monocarboxylate transporter 1 (MCT1) plays a vital role in oligodendrocyte maturation and myelin synthesis. Herein, we hypothesized that miR-219 might promote oligodendrocyte differentiation and attenuate demyelination in a cuprizone (CPZ)-induced demyelinated model by regulating the expression of MCT1. We found that CPZ-treated mice exhibited significantly increased anxiety in the open field test. However, miR-219 reduced anxiety as shown by an increase in the total distance, the central distance and the mean amount of time spent in the central area. miR-219 decreased the quantity of OPCs and increased the number of oligodendrocytes and the level of myelin basic protein (MBP) and cyclic nucleotide 3' phosphodiesterase (CNP) protein. Ultrastructural studies further confirmed that the extent of demyelination was attenuated by miR-219 overexpression. Meanwhile, miR-219 also greatly enhanced MCT1 expression via suppression of oligodendrocyte differentiation inhibitors, Sox6 and Hes5, treatment with the MCT1 inhibitor α-cyano-4-hydroxycinnamate (4-CIN) reduced the number of oligodendrocytes and the protein levels of MBP and CNP. Taken together, these results suggest a novel mode of action of miR-219 via MCT1 in vivo and may provide a new potential remyelination therapeutic target.


Subject(s)
Coumaric Acids/pharmacology , Cuprizone/pharmacology , Demyelinating Diseases/drug therapy , MicroRNAs/genetics , Monocarboxylic Acid Transporters/metabolism , Oligodendroglia/drug effects , Symporters/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Corpus Callosum/metabolism , Demyelinating Diseases/genetics , Mice, Inbred C57BL , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Stem Cells/classification , Stem Cells/metabolism
9.
Biochem Biophys Res Commun ; 491(3): 636-641, 2017 09 23.
Article in English | MEDLINE | ID: mdl-28757412

ABSTRACT

Aging is accompanied by deficits in cognitive function and neuronal degeneration or loss. Quercetin is a flavonoid that exhibits powerful antioxidant activity. This study evaluated the protective effects and mechanisms of quercetin in d-galactose-induced neurotoxicity in mice. Quercetin was administered daily at doses of 20 or 50 mg/kg in d-galactose-injected (50 mg/kg/subcutaneous (s.c.)) mice for eight weeks. Morris water maze tests demonstrated that quercetin significantly improved learning and memory compared to d-galactose-treated control mice. Quercetin also prevented changes in the neuronal cell morphology and apoptosis in the hippocampus as well as increased the expression of Nrf2, HO-1 and SOD in d-galactose-treated mice. Treatment with the Nrf2 inhibitor Brusatol reversed the effects of quercetin on HO-1 and SOD expression as well as neuronal cell protection. In conclusion, quercetin protected mice from d-galactose-induced cognitive functional impairment and neuronal cell apoptosis via activation of the Nrf2-ARE signaling pathway.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Cognition Disorders/prevention & control , Cognition Disorders/physiopathology , Hippocampus/drug effects , NF-E2-Related Factor 2/metabolism , Quercetin/pharmacology , Animals , Antioxidants/administration & dosage , Antioxidants/pharmacology , Cognition Disorders/chemically induced , Dose-Response Relationship, Drug , Galactose , Hippocampus/pathology , Hippocampus/physiopathology , Learning Disabilities/chemically induced , Learning Disabilities/physiopathology , Learning Disabilities/prevention & control , Male , Memory Disorders/chemically induced , Memory Disorders/physiopathology , Memory Disorders/prevention & control , Mice , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Neurotoxins , Quercetin/administration & dosage , Signal Transduction/drug effects , Treatment Outcome
10.
Cell Mol Neurobiol ; 37(4): 743-752, 2017 May.
Article in English | MEDLINE | ID: mdl-27456026

ABSTRACT

In this study, we investigated the neuroprotective effect of Ro25-6981 against cerebral ischemia/reperfusion injury. Ro25-6981 alone or in combination with rapamycin was intracerebroventricularly administered to rats which suffered transient forebrain ischemia inducing by 4-vessel occlusion and reperfusion. Nissl staining was used to determine the survival of CA1 pyramidal cells of the hippocampus, while immunohistochemistry was performed to measure neuron-specific enolase (NSE) expression. The expression of autophagy-related proteins, such as microtubule-associated protein l light chain 3 (LC3), Beclin 1, and sequestosome 1 (p62), was assessed by immunoblotting. Nissl staining showed that neuronal damage was reduced in the hippocampal CA1 pyramidal layer in rats that received Ro25-6981. The protective effect of Ro25-6981 was dose-dependent, with a significant effect in the middle-dose range. The expression of NSE increased after Ro25-6981 treatment. Ro25-6981 significantly decreased LC3II (which is membrane bound) and Beclin 1, and increased p62. In addition, Ro25-6981 decreased rapamycin-induced neuronal damage and excessive activation of autophagy after I/R. Taken together, the results suggest that Ro25-6981 could suppress ischemic brain injury by regulating autophagy-related proteins during ischemia/reperfusion.


Subject(s)
Autophagy/drug effects , Brain Injuries/drug therapy , Neuroprotective Agents/pharmacology , Phenols/pharmacology , Piperidines/pharmacology , Reperfusion Injury/drug therapy , Animals , Apoptosis Regulatory Proteins/metabolism , Brain/metabolism , Brain Ischemia/drug therapy , Disease Models, Animal , Male , Microtubule-Associated Proteins/metabolism , Neuroprotection/drug effects , Rats, Sprague-Dawley , Reperfusion Injury/complications , Reperfusion Injury/prevention & control
11.
Brain Behav Immun ; 57: 193-199, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27006279

ABSTRACT

T helper cells 17 (Th17) are recognized as key participants in the pathogenesis of chronic autoimmune diseases such as multiple sclerosis (MS). Regulation of Th17 differentiation is a valuable strategy for diagnosis and treatment of these complicated immune disorders. Here, by genome-wide expression profiling of microRNAs (miRs), we screened miR-30a, whose level was greatly decreased during Th17 differentiation and the process of demyelination disease, both in MS patients and experimental autoimmune encephalomyelitis (EAE) mice. Enforced constitutive expression of miR-30a in naïve T cells inhibited their differentiation into Th17, and in vivo overexpression of miR-30a resulted in fewer Th17 and alleviative EAE. Moreover, target prediction analysis and dual luciferase report assay revealed that interleukin-21 receptor (IL-21R) was a direct target of miR-30a, a finding consistent with the results that miR-30a downregulated the expression of IL-21R, while overexpression of IL-21R alleviated the inhibitory effect of miR-30a on Th17 differentiation. Taken together, our findings imply that miR-30a inhibits Th17 differentiation and the pathogenesis of MS by targeting IL-21R.


Subject(s)
Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/metabolism , Interleukin-21 Receptor alpha Subunit/metabolism , MicroRNAs/metabolism , Multiple Sclerosis/metabolism , Th17 Cells , Adult , Animals , Humans , Mice , Mice, Inbred C57BL
12.
Biochim Biophys Acta ; 1839(11): 1161-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25201174

ABSTRACT

Abnormally high transcription of the glial cell-line derived neurotrophic factor (gdnf) gene in glioma cells is related to the hyperacetylation of histone H3 lysine 9 (H3K9) in its promoter region II, but the mechanism remains unclear. There are three consecutive putative binding sites for the transcription factor early growth response protein 1(Egr-1) in promoter region II of the gdnf gene, and Egr-1 participates in gdnf gene transcription activation. Here we show that the acetylation level of H3K9 at Egr-1 binding sites in gdnf gene promoter region II in rat C6 astroglioma cells was significantly higher than that in normal astrocytes, and the binding capacity was also significantly higher. In C6 astroglioma cells, gdnf gene transcription significantly decreased after Egr-1 knock-down. In addition, the deletion or mutation of the Egr-1 binding site also significantly down-regulated the activity of promoter region II of this gene in vitro. When curcumin decreased the acetylation level of H3K9 at the Egr-1 binding site, the binding of Egr-1 to promoter region II and GDNF mRNA levels significantly decreased. In contrast, trichostatin A treatment significantly increased H3K9 acetylation at the Egr-1 binding site, which significantly increased both the binding of Egr-1 with promoter region II and GDNF mRNA levels. In this context, knocking down Egr-1 significantly reduced the elevation in gdnf gene transcription. Collectively, our results demonstrate that the hyperacetylation of H3K9 at Egr-1 binding sites in promoter region II of the gdnf gene can up-regulate the binding of Egr-1 to increase gdnf gene transcription in glioma cells.


Subject(s)
Early Growth Response Protein 1/physiology , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glioma/genetics , Glioma/metabolism , Histone Acetyltransferases/metabolism , Histones/metabolism , Acetylation , Animals , Binding Sites , Cells, Cultured , Gene Expression Regulation, Neoplastic , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Promoter Regions, Genetic , Rats
13.
Stem Cells ; 32(7): 1789-804, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24648391

ABSTRACT

GABAergic interneurons regulate cortical neural networks by providing inhibitory inputs, and their malfunction, resulting in failure to intricately regulate neural circuit balance, is implicated in brain diseases such as Schizophrenia, Autism, and Epilepsy. During early development, GABAergic interneuron progenitors arise from the ventral telencephalic area such as medial ganglionic eminence (MGE) and caudal ganglionic eminence (CGE) by the actions of secreted signaling molecules from nearby organizers, and migrate to their target sites where they form local synaptic connections. In this study, using combinatorial and temporal modulation of developmentally relevant dorsoventral and rostrocaudal signaling pathways (SHH, Wnt, and FGF8), we efficiently generated MGE cells from multiple human pluripotent stem cells. Most importantly, modulation of FGF8/FGF19 signaling efficiently directed MGE versus CGE differentiation. Human MGE cells spontaneously differentiated into Lhx6-expressing GABAergic interneurons and showed migratory properties. These human MGE-derived neurons generated GABA, fired action potentials, and displayed robust GABAergic postsynaptic activity. Transplantation into rodent brains results in well-contained neural grafts enriched with GABAergic interneurons that migrate in the host and mature to express somatostatin or parvalbumin. Thus, we propose that signaling modulation recapitulating normal developmental patterns efficiently generate human GABAergic interneurons. This strategy represents a novel tool in regenerative medicine, developmental studies, disease modeling, bioassay, and drug screening.


Subject(s)
Brain/cytology , Interneurons/physiology , Pluripotent Stem Cells/physiology , Animals , Body Patterning , Brain/embryology , Cell Line , Fibroblast Growth Factors/physiology , GABAergic Neurons/physiology , Hedgehog Proteins/metabolism , Humans , Interneurons/transplantation , Mice , Mice, Inbred NOD , Mice, SCID , Neural Stem Cells/physiology , Signal Transduction
14.
Cell Mol Neurobiol ; 35(7): 913-20, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25833395

ABSTRACT

One of the pathological hallmarks of periventricular white matter injury is the vulnerability of pre-oligodendrocytes (preOLs) to hypoxia-ischemia (HI). There is increasing evidence that basic fibroblast growth factor (bFGF) is an important signaling molecule for neurogenesis and neuroprotection in the central nervous system. However, it is unknown whether bFGF protects preOLs from oxygen/glucose deprivation (OGD) damage in vitro and promotes remyelination in HI-induced rats. In this present study, bFGF exerted a protective effect on myelin by increasing the myelin thickness, the number of myelinated axons, and myelin basic protein expression in the HI-induced demyelinated neonatal rat corpus callosum. In vitro, bFGF ameliorated the impaired mitochondria and cell processes induced by OGD to promote the survival of isolated O4-positive preOLs. Additionally, the expression of fibroblast growth factor receptor 3 (FGFR3) was dramatically up-regulated in the preOLs after bFGF administration in vivo and in vitro. Thus, bFGF-stimulated remyelination in HI-induced rats by protecting the preOLs from hypoxic injury, and the mechanism involved may be mediated by FGFR3.


Subject(s)
Demyelinating Diseases/drug therapy , Fibroblast Growth Factor 2/therapeutic use , Glucose/deficiency , Neural Stem Cells/drug effects , Oligodendroglia/drug effects , Oxygen/metabolism , Animals , Animals, Newborn , Cells, Cultured , Demyelinating Diseases/metabolism , Fibroblast Growth Factor 2/pharmacology , Male , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurogenesis/physiology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oligodendroglia/metabolism , Rats , Rats, Sprague-Dawley
15.
Dev Growth Differ ; 56(7): 511-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25200136

ABSTRACT

Oligodendrocyte progenitor cells (OPCs) transplantation is receiving considerable attention in the field of regenerative medicine therapy for demyelinating diseases. Although embryonic stem cells (ESCs) have been successfully induced to differentiate into OPCs with cytokines cocktails in vitro, the regulatory roles of many key transcription factors in this process are not clear. Here, we introduced oligodendrocyte lineage transcription factor 2 (Olig2), a basic helix-loop-helix transcription factor, into mouse embryonic stem cells (mESCs) to investigate its effects on the differentiation of mESCs into OPCs. The results showed that Olig2 overexpression alone did not affect pluripotency of mESCs, but in the stimulation of differentiating cocktails, Olig2 accelerated mESCs to differentiate into OPCs, shortening the induction time span from normal 21 days to 11 days. Further study demonstrated the Olig2-mESCs derived OPCs were able to differentiate into C-type natriuretic peptid (CNP) and Myelin Basic Protein (MBP) positive mature oligodendrocytes (OLs) in vitro, suggesting these induced OPCs might be favorable for myelin regeneration in vivo.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Nerve Tissue Proteins/metabolism , Oligodendroglia/cytology , Stem Cells/cytology , Analysis of Variance , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Cell Differentiation/genetics , Fluorescent Antibody Technique , Mice , Nerve Tissue Proteins/genetics , Oligodendrocyte Transcription Factor 2 , Regenerative Medicine/methods
16.
Cell Mol Neurobiol ; 34(3): 463-71, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24519463

ABSTRACT

The aim of this study was to investigate quercetin's (Qu) ability to promote proliferation and differentiation of oligodendrocyte precursor cells (OPCs) under oxygen/glucose deprivation (OGD)-induced injury in vitro. The results showed that after OGD, OPCs survival rate was significantly increased by Qu as measured by Cell Counting Kit-8. Furthermore, Qu treatment reduced apoptosis of OPCs surveyed by Hoechst 33258 nuclear staining. Qu at 9 and 27 µM promoted the proliferation of OPCs the most by Brdu and Olig2 immunocytochemical staining after OGD 3 days. Also, Qu treatment for 8 days after OGD, the differentiation of OPCs to oligodendrocyte was detected by immunofluorescence staining showing that O4, Olig2, and myelin basic protein (MBP) positive cells were significantly increased compared to control group. Additionally, the protein levels of Olig2 and MBP of OPCs were quantified using western blot and mRNA levels of Olig2 and Inhibitor of DNA binding 2 (Id2) were measured by RT-PCR. Western blot showed a significant increase in Olig2 and MBP expression levels compared with controls after OGD and Qu treatment with a linear does-response curve from 3 to 81 µM. After treatment with Qu compared to its control group, Olig2 mRNA level was significantly up-regulated, whereas Id2 mRNA level was down-regulated. In conclusion, Qu at 3-27 µM can promote the proliferation and differentiation of OPCs after OGD injury and may regulate the activity of Olig2 and Id2.


Subject(s)
Cell Differentiation/physiology , Glucose/metabolism , Oligodendroglia/metabolism , Oxygen/metabolism , Quercetin/pharmacology , Stem Cells/metabolism , Animals , Animals, Newborn , Antioxidants/pharmacology , Cell Differentiation/drug effects , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Glucose/deficiency , Humans , Oligodendroglia/drug effects , Rats , Rats, Sprague-Dawley , Stem Cells/drug effects
17.
Int J Biol Macromol ; 278(Pt 1): 134694, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39142476

ABSTRACT

The wearability of the flexible electronic skin (e-skin) allows it to attach to the skin for human motion monitoring, which is essential for studying human motion and especially for assessing how well patients are recovering from rehabilitation therapy. However, the use of non-degradable synthetic materials in e-skin may raise skin safety concerns. Natural biodegradable polymers with advantages such as biodegradability, biocompatibility, sustainability, natural abundance, and low cost have the potential to be alternative materials for constructing flexible e-skin and applying them to human motion monitoring. This review summarizes the applications of natural biodegradable polymers in e-skin for human motion monitoring over the past three years, focusing on the discussion of cellulose, chitosan, silk fibroin, gelatin, and sodium alginate. Finally, we summarize the opportunities and challenges of e-skin based on natural biodegradable polymers. It is hoped that this review will provide insights for the future development of flexible e-skin in the field of human motion monitoring.

18.
Biochem Mol Biol Educ ; 52(3): 323-331, 2024.
Article in English | MEDLINE | ID: mdl-38308542

ABSTRACT

The primary objective of science postgraduate education is to foster students' capacity for creative thinking and problem-solving, particularly in the context of scientific research quality. In order to achieve this goal, the "7E" teaching mood has been implemented in the cell biology course for postgraduate students to promote student-centered active inquiry learning instead of breaking away from traditional indoctrination-based teaching methods. This study demonstrates that the implementation of the "7E" teaching mode, through content programming, process design, and effect evaluation, effectively meets the needs of the majority of students, fosters their interest in learning, enhances their performance in comprehensive questioning, and enhances their innovative abilities in scientific research. Consequently, this research offers a theoretical framework and practical foundation for the development of the "7E" teaching mode in postgraduate courses, aiming to cultivate highly skilled scientific professionals.


Subject(s)
Cell Biology , Problem-Based Learning , Students , Humans , Students/psychology , Problem-Based Learning/methods , Cell Biology/education , Teaching , Curriculum , Education, Graduate/methods , Learning
19.
Phytomedicine ; 124: 155280, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38183697

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is the most common reproductive-endocrine condition in premenopausal women. Troxerutin, a common clinical anti-coagulant agent, was shown to work as a strong IL-22 boosting agent counteracting the hyperactivated gonadotrophin releasing hormone (GnRH) neurons and heightened GnRH release, the neuroendocrine origin of PCOS with unknown mechanism in rats. Exploring the off-label use of troxerutin medication for PCOS is thus sorely needed. METHODS: Serum IL-22 content and hypothalamic IL-22 protein were detected. Inflammatory factor levels in hypothalamo-pituitary were evaluated. Immunofluorescence staining was employed to determine the activation and M1/M2-prone polarization of microglia in arcuate hypothalamus and median eminence. RNA-sequencing and transcriptome analysis were applied to explore the potential driver of microglia M2-polarization in response to IL-22 bolstering effect. The function of microglial IL-22/IL-22R1/IRF3 system was further verified using in vivo knockdown of IL-22R1 and a potent IRF3 inhibitor in BV2 microglial cell lines in vitro. RESULTS: Troxerutin augmented serum IL-22 content, and its consequent spillover into the hypothalamus led to the direct activation of IL-22R1/IRF3 system on microglia, thereby promoted microglia M2 polarization in arcuate hypothalamus and median eminence, dampened hypothalamic neuroinflammation, inhibited hyperactive GnRH and rescued a breadth of PCOS-like traits in dihydrotestosterone (DHT) rats. The salutary effects of troxerutin treatment on hypothalamic neuroinflammation, microglial M1/2 polarization, GnRH secretion and numerous PCOS-like features were blocked by in vivo knockdown of IL-22R1. Moreover, evidence in vitro illustrated that IL-22 supplement to BV-2 microglia cell lines promoted M2 polarization, overproduction of anti-inflammatory marker and limitation of pro-inflammatory factors, whereas these IL-22 effects were blunted by geldanamycin, a potent IRF3 inhibitor. CONCLUSION: Here, the present study reported the potential off-label use of troxerutin medication, a common clinical anti-coagulant agent and an endogenous IL-22 enhancer, for multiple purposes in PCOS. The rational underlying the application of troxerutin as a therapeutic choice in PCOS derived from its activity as an IL-22 memetic agent targeting the neuro-endocrine origin of PCOS, and its promotive impact on microglia M2 polarization via activating microglial IL-22R1/IRF3 system in the arcuate hypothalamus and median eminence of DHT female rats.


Subject(s)
Hydroxyethylrutoside/analogs & derivatives , Polycystic Ovary Syndrome , Receptors, Interleukin , Humans , Rats , Female , Animals , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/drug therapy , Dihydrotestosterone/adverse effects , Dihydrotestosterone/metabolism , Microglia , Neuroinflammatory Diseases , Interleukin-22 , Hypothalamus/metabolism , Gonadotropin-Releasing Hormone/adverse effects , Gonadotropin-Releasing Hormone/metabolism , Interferon Regulatory Factor-3/metabolism
20.
Neurol Res ; : 1-12, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38979727

ABSTRACT

OBJECTIVES: This study aims to investigate the role of high-intensity interval training (HIIT) in promoting myelin sheath recovery during the remyelination phase in cuprizone (CPZ)-induced demyelination mice and elucidate the mechanisms involving the Wnt/ß-catenin pathway. METHODS: After 5 weeks of a 0.2% CPZ diet to induce demyelination, a 4-week recovery phase with a normal diet was followed by HIIT intervention. Mice body weight was monitored. Morris water maze (MWM) gauged spatial cognition and memory, while the open field test (OFT) assessed anxiety levels. Luxol fast blue (LFB) staining measured demyelination, and immunofluorescence examined myelin basic protein (MBP) and platelet-derived growth factor receptor-alpha (PDGFR-α). Western blotting analyzed protein expression, including MBP, PDGFR-α, glycogen synthase kinase-3ß (GSK3ß), ß-catenin, and p-ß-catenin. Real-time PCR detected mRNA expression levels of CGT and CST. RESULTS: HIIT promoted remyelination in demyelinating mice, enhancing spatial cognition, memory, and reducing anxiety. LFB staining indicated decreased demyelination in HIIT-treated mice. Immunofluorescence demonstrated increased MBP fluorescence intensity and PDGFR-α+ cell numbers with HIIT. Western blotting revealed HIIT reduced ß-catenin levels while increasing p-ß-catenin and GSK3ß levels. Real-time PCR demonstrated that HIIT promoted the generation of new myelin sheaths. Additionally, the Wnt/ß-catenin pathway agonist, SKL2001, decreased MBP expression but increased PDGFR-α expression. DISCUSSION: HIIT promotes remyelination by inhibiting the Wnt/ß-catenin pathway and is a promising rehabilitation training for demyelinating diseases. It provides a new theoretical basis for clinical rehabilitation and care programs.

SELECTION OF CITATIONS
SEARCH DETAIL