Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Odontology ; 112(4): 1266-1273, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38393516

ABSTRACT

Nuclear factor kappa-B (NF-κB) signaling-mediated inflammation contributes greatly to the pathogenesis of periodontitis. Neddylation, a ubiquitin-like posttranslational modification, is known to regulate NF-κB signaling. DCUN1D1 (defective in cullin neddylation 1 domain containing 1) is a critical factor in neddylation and has been shown to regulate NF-κB activation. However, the previse roles of DCUN1D1 in periodontitis are not fully elucidated. To explore the roles of DCUN1D1 in periodontitis, the expression of DCUN1D1 was measured in gingival tissues of patients with periodontitis. We inhibited DCUN1D1 by siRNA knocking down or using inhibitor in gingival fibroblasts and the lipopolysaccharides (LPS)-induced expression of IL-6 and TNF-α, and activation of NF-κB were measured. The expression of DCUN1D1 was increased in gingival tissues of patients with periodontitis. Knocking down or inhibiting DCUN1D1 suppressed LPS-induced production of IL-6 and TNF-α, decreased NF-κB activity, and inhibited LPS-induced activation of NF-κB. Inhibiting DCUN1D1 ameliorates periodontitis by suppressing NF-κB signaling.


Subject(s)
Interleukin-6 , Lipopolysaccharides , NF-kappa B , Periodontitis , Signal Transduction , Tumor Necrosis Factor-alpha , NF-kappa B/metabolism , Periodontitis/metabolism , Humans , Interleukin-6/metabolism , Tumor Necrosis Factor-alpha/metabolism , RNA, Small Interfering , Fibroblasts/metabolism , Gingiva/metabolism , Blotting, Western , Cells, Cultured , Real-Time Polymerase Chain Reaction , Male , Enzyme-Linked Immunosorbent Assay
2.
J Nanobiotechnology ; 21(1): 172, 2023 May 30.
Article in English | MEDLINE | ID: mdl-37248505

ABSTRACT

Engineered nanosystems offer a promising strategy for macrophage-targeted therapies for various diseases, and their physicochemical parameters including surface-active ligands, size and shape are widely investigated for improving their therapeutic efficacy. However, little is known about the synergistic effect of elasticity and surface-active ligands. Here, two kinds of anti-inflammatory N-acetylcysteine (NAC)-loaded macrophage-targeting apoptotic-cell-inspired phosphatidylserine (PS)-containing nano-liposomes (PSLipos) were constructed, which had similar size and morphology but different Young's modulus (E) (H, ~ 100 kPa > Emacrophage vs. L, ~ 2 kPa < Emacrophage). Interestingly, these PSLipos-NAC showed similar drug loading and encapsulation efficiency, and in vitro slow-release behavior of NAC, but modulus-dependent interactions with macrophages. Softer PSLipos-L-NAC could resist macrophage capture, but remarkably prolong their targeting effect period on macrophages via durable binding to macrophage surface, and subsequently more effectively suppress inflammatory response in macrophages and then hasten inflammatory lung epithelial cell wound healing. Especially, pulmonary administration of PSLipos-L-NAC could significantly reduce the inflammatory response of M1-like macrophages in lung tissue and promote lung injury repair in a bleomycin-induced acute lung injury (ALI) mouse model, providing a potential therapeutic approach for ALI. The results strongly suggest that softness may enhance ligand-directed macrophage-mediated therapeutic efficacy of nanosystems, which will shed new light on the design of engineered nanotherapeutics.


Subject(s)
Acute Lung Injury , Lung , Mice , Animals , Lung/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Macrophages/metabolism , Acetylcysteine/metabolism , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use
3.
Odontology ; 111(2): 474-486, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36181561

ABSTRACT

It has been demonstrated that circular RNA (circRNA) is involved in the progression of tongue squamous cell carcinoma (TSCC). The aim of this study was to investigate the intrinsic mechanism of circ_0081069 in TSCC progression. The expression levels of circ_00081069, miR-634, and mitogen-activated protein kinase kinase 4 (MAP2K4) in TSCC tissues and cells were detected by quantitative real-time PCR (qRT-PCR). Cell counting kit 8 assay, Edu assay, and flow cytometry assay were used to detect cell proliferation and cell cycle distribution. Transwell assay was used to detect cell migration and invasion abilities. Western blot analysis was performed to detect the protein expression. Dual-luciferase reporter assay was used to detect the targeting relationships of circ_0081069, miR-634 and MAP2K4. Immunohistochemical staining was used to measure MAP2K4-positive cells in tissues. The effect of circ_0081069 silencing on tumor formation in TSCC in vivo was explored by xenograft tumor assay. Circ_0081069 was highly expressed in TSCC tissues and cells. Silencing of circ_0081069 inhibited cell proliferation, cell cycle progress, cell migration and invasion in vitro, as well as hindered tumor growth in vivo. Mechanistically, circ_0081069 targeted miR-634 to negatively regulate miR-634 expression, and inhibition of miR-634 was able to weaken the inhibitory effect of circ_0081069 knockdown on proliferation, migration, and invasion of TSCC cells. MiR-634 targeted MAP2K4 and negatively regulated MAP2K4 expression, and overexpression of miR-634 inhibited TSCC cell proliferation, migration, and invasion, while co-overexpression of MAP2K4 was able to reverse the effects of miR-634 in TSCC cells. Circ_0081069 is involved in the regulation of proliferation, cycle progress, migration, and invasion of TSCC cells through the miR-634/MAP2K4 axis and has the potential to serve as a diagnostic biomarker and therapeutic target.


Subject(s)
Carcinoma, Squamous Cell , MicroRNAs , Tongue Neoplasms , Humans , Carcinoma, Squamous Cell/genetics , Tongue Neoplasms/genetics , Cell Movement , Cell Proliferation , Tongue , MicroRNAs/genetics , Cell Line, Tumor , MAP Kinase Kinase 4
4.
Future Oncol ; 18(2): 215-230, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34854737

ABSTRACT

Aims: This study presents a survival stratification model based on multi-omics integration using bidirectional deep neural networks (BiDNNs) in gastric cancer. Methods: Based on the survival-related representation features yielded by BiDNNs through integrating transcriptomics and epigenomics data, K-means clustering analysis was performed to cluster tumor samples into different survival subgroups. The BiDNNs-based model was validated using tenfold cross-validation and in two independent confirmation cohorts. Results: Using the BiDNNs-based survival stratification model, patients were grouped into two survival subgroups with log-rank p-value = 9.05E-05. The subgroups classification was robustly validated in tenfold cross-validation (C-index = 0.65 ± 0.02) and in two confirmation cohorts (E-GEOD-26253, C-index = 0.609; E-GEOD-62254, C-index = 0.706). Conclusion: We propose and validate a robust and stable BiDNN-based survival stratification model in gastric cancer.


Subject(s)
Biomarkers, Tumor/genetics , Stomach Neoplasms/mortality , Unsupervised Machine Learning , Aged , Cluster Analysis , DNA Methylation , Datasets as Topic , Epigenomics/methods , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Male , Middle Aged , RNA-Seq/methods , Risk Assessment/methods , Stomach Neoplasms/genetics
5.
Mol Biol Evol ; 37(8): 2300-2308, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32243529

ABSTRACT

Fisher's fundamental theorem of natural selection predicts no additive variance of fitness in a natural population. Consistently, studies in a variety of wild populations show virtually no narrow-sense heritability (h2) for traits important to fitness. However, counterexamples are occasionally reported, calling for a deeper understanding on the evolution of additive variance. In this study, we propose adaptive divergence followed by population admixture as a source of the additive genetic variance of evolutionarily important traits. We experimentally tested the hypothesis by examining a panel of ∼1,000 yeast segregants produced by a hybrid of two yeast strains that experienced adaptive divergence. We measured >400 yeast cell morphological traits and found a strong positive correlation between h2 and evolutionary importance. Because adaptive divergence followed by population admixture could happen constantly, particularly in species with wide geographic distribution and strong migratory capacity (e.g., humans), the finding reconciles the observation of abundant additive variances in evolutionarily important traits with Fisher's fundamental theorem of natural selection. Importantly, the revealed role of positive selection in promoting rather than depleting additive variance suggests a simple explanation for why additive genetic variance can be dominant in a population despite the ubiquitous between-gene epistasis observed in functional assays.


Subject(s)
Adaptation, Biological , Biological Evolution , Genetic Fitness , Selection, Genetic , Saccharomyces cerevisiae
6.
J Cell Mol Med ; 24(13): 7313-7330, 2020 07.
Article in English | MEDLINE | ID: mdl-32441057

ABSTRACT

Epidermal growth factor (EGF) has many physiological roles. However, its effects on stem and progenitor Leydig cell development remain unclear. Rat stem and progenitor Leydig cells were cultured with different concentrations of EGF alone or in combination with EGF antagonist, erlotinib or cetuximab. EGF (1 and 10 ng/mL) stimulated the proliferation of stem Leydig cells on the surface of seminiferous tubules and isolated CD90+ stem Leydig cells and progenitor Leydig cells but it blocked their differentiation. EGF also exerted anti-apoptotic effects of progenitor Leydig cells. Erlotinib and cetuximab are able to reverse EGF-mediated action. Gene microarray and qPCR of EGF-treated progenitor Leydig cells revealed that the down-regulation of steroidogenesis-related proteins (Star and Hsd3b1) and antioxidative genes. It was found that EGF acted as a proliferative agent via increasing phosphorylation of AKT1. In conclusion, EGF stimulates the proliferation of rat stem and progenitor Leydig cells but blocks their differentiation.


Subject(s)
Epidermal Growth Factor/pharmacology , Leydig Cells/cytology , Stem Cells/cytology , Stem Cells/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Lineage/drug effects , Cell Lineage/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , ErbB Receptors/metabolism , Gene Expression Regulation/drug effects , Leydig Cells/drug effects , Leydig Cells/metabolism , Male , Phosphorylation/drug effects , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Stem Cells/drug effects , Steroids/biosynthesis , Thymidine/metabolism
7.
Fish Shellfish Immunol ; 97: 509-514, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31877360

ABSTRACT

In this study, the effect of Lycium barbarum polysaccharides (LBP) on immunological parameters, apoptosis, and growth performance of Nile tilapia (Oreochromis niloticus) was investigated. Dietary supplementation with LBP significantly increased complement 3 (C3) activity and promoted interleukin IL-1ß gene expression in spleen tissue, significantly reduced apoptosis in spleen tissue, increased the specific growth rate (SGR), relative length gain (LG), and relative weight gain (WG) of Nile tilapia. However, dietary supplementation with LBP did not have a significant effect on serum alkaline phosphatase (AKP), malondialdehyde (MDA), and superoxide dismutase (SOD), blood constituents, apoptosis, or gene expression of IL-1ß in liver tissue. Overall, the results showed that dietary supplementation with LBP increased the nonspecific immunity of Nile tilapia and reduced the apoptosis rate to promote growth and development. Thus, LBP has potential for use as a new immunostimulant in aquaculture.


Subject(s)
Apoptosis/drug effects , Cichlids/growth & development , Cichlids/immunology , Dietary Supplements , Drugs, Chinese Herbal/administration & dosage , Animal Feed , Animals , Aquaculture , Complement C3/immunology , Fish Diseases/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology
8.
Stem Cells ; 35(5): 1222-1232, 2017 05.
Article in English | MEDLINE | ID: mdl-28090714

ABSTRACT

Stem Leydig cell (SLC) transplantation could provide a new strategy for treating the testosterone deficiency. Our previous study demonstrated that CD51 (also called integrin αv) might be a putative cell surface marker for SLCs, but the physiological function and efficacy of CD51+ SLCs treatment remain unclear. Here, we explore the potential therapeutic benefits of CD51+ SLCs transplantation and whether these transplanted cells can be regulated by the hypothalamic-pituitary-gonadal (HPG) axis. CD51+ cells were isolated from the testes of 12-weeks-old C57BL/6 mice, and we showed that such cells expressed SLC markers and that they were capable of self-renewal, extensive proliferation, and differentiation into multiple mesenchymal cell lineages and LCs in vitro. As a specific cytotoxin that eliminates Leydig cells (LCs) in adult rats, ethane dimethanesulfonate (EDS) was used to ablate LCs before the SLC transplantation. After being transplanted into the testes of EDS-treated rats, the CD51+ cells differentiated into mature LCs, and the recipient rats showed a partial recovery of testosterone production and spermatogenesis. Notably, a testosterone analysis revealed a circadian rhythm of testosterone secretion in cell-transplanted rats, and these testosterone secretions could be suppressed by decapeptyl (a luteinizing hormone-releasing hormone agonist), suggesting that the transplanted cells might be regulated by the HPG axis. This study is the first to demonstrate that CD51+ SLCs can restore the neuroendocrine regulation of testicular function by physiologically recovering the expected episodic changes in diurnal testosterone serum levels and that SLC transplantation may provide a new tool for the studies of testosterone deficiency treatment. Stem Cells 2017;35:1222-1232.


Subject(s)
Integrin alphaV/metabolism , Leydig Cells/cytology , Stem Cell Transplantation , Stem Cells/cytology , Testosterone/deficiency , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Lineage , Cell Self Renewal , Cell Separation , Disease Models, Animal , Hypothalamo-Hypophyseal System/metabolism , Male , Mesylates , Mice, Inbred C57BL , Organ Size , Rats, Sprague-Dawley , Spermatogenesis , Testis/cytology
9.
Mol Cell Neurosci ; 71: 80-91, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26711806

ABSTRACT

The Nogo-66 receptor (NgR1), a receptor for Nogo-A, contributes to the inhibition of axonal regeneration in the adult central nervous system after traumatic injuries. Thus, NgR1 has been considered a critical target in axon regeneration therapy. Here, we identified a specific NgR1 antagonist peptide (HIYTALV, named NAP2) which promotes neurite regeneration in vitro from a phage display heptapeptide library. NAP2 was co-localized with NgR1 on the surface of PC12 cells and cerebellar granule cells (CGCs) by immunofluorescence assay. Horseradish peroxidase (HRP)-streptavidin-biotin assay further showed that NAP2 binds to NgR1 and the dissociation constant (Kd) was 0.45 µM Functional analyses indicated that NAP2 could reduce the inhibitory effects of Nogo-66 on neurite outgrowth in differentiated PC12 cells and CGCs by blocking the Nogo-66-induced activation of Rho-associated coiled coil-containing protein kinase (ROCK), collapsin response mediator protein 2 (CRMP2) and myosin light chain (MLC). Taken together, the small molecule NgR1 antagonist peptide NAP2 (MW: 815.98Da) has a potential ability in crossing blood brain barrier and will be a promising therapeutic agent for the treatment of spinal cord injury and neurodegenerative diseases.


Subject(s)
Myelin Proteins/antagonists & inhibitors , Nerve Regeneration , Neurites/drug effects , Oligopeptides/pharmacology , Animals , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Ligands , Myelin Proteins/metabolism , Myosin Light Chains/metabolism , Nerve Tissue Proteins/metabolism , Neurites/metabolism , Neurites/physiology , Nogo Proteins , PC12 Cells , Protein Binding , Rats , Rats, Sprague-Dawley , rho-Associated Kinases/metabolism
10.
Mar Drugs ; 15(6)2017 Jun 03.
Article in English | MEDLINE | ID: mdl-28587208

ABSTRACT

The goal of this study was to examine the effects of xyloketal B on nonalcoholic fatty liver disease (NAFLD) and to explore the molecular mechanisms underlying its effects in both in vivo and in vitro models. We discovered an association between xyloketal B and the sterol regulatory element-binding protein-1c (SREBP-1c) signaling pathway, which is related to lipid metabolism. Mice were dosed with xyloketal B (5, 10 and 20 mg/kg/d) and atorvastatin (15 mg/kg/d) via intraperitoneal injection once daily for 40 days after being fed a high fat diet plus 10% high fructose liquid (HFD+HFL) for 8 weeks. Xyloketal B significantly improved HFD+HFL-induced hepatic histological lesions and attenuated lipid and glucose accumulation in the blood as well as lipid accumulation in the liver. Xyloketal B increased the expression of CPT1A, and decreased the expression of SREBP-1c and its downstream targeting enzymes such as ACC1, ACL, and FAS. Xyloketal B also significantly reduced lipid accumulation in HepG2 cells treated with free fatty acids (FFAs). These data suggested that xyloketal B has lipid-lowering effects via the SREBP-1c pathway that regulate lipid metabolism. Thus, targeting SREBP-1c activation with xyloketal B may be a promising novel approach for NAFLD treatment.


Subject(s)
Fatty Acids/metabolism , Lipid Metabolism/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Pyrans/pharmacology , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Cell Line, Tumor , Diet, High-Fat/adverse effects , Fructose/metabolism , Hep G2 Cells , Humans , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects
11.
Growth Factors ; 34(5-6): 166-176, 2016 12.
Article in English | MEDLINE | ID: mdl-27760485

ABSTRACT

Leukemia inhibitory factor (LIF) has many physiological roles. However, its effects on Leydig cell development are still unclear. Rat immature and adult Leydig cells were cultured with different concentrations of LIF alone or in combination with luteinizing hormone (LH) for 24 h. LIF (1 and 10 ng/ml) significantly increased androgen production in immature Leydig cells, but had no effects on testosterone production in adult Leydig cells. Further studies revealed that LIF dose-dependently increased Star and Hsd17b3 expression levels in immature Leydig cells. Gene microarray revealed that the upregulation of anti-oxidative genes and Star might contribute to LIF-induced androgen production. In conclusion, LIF has stimulatory effects on androgen production in rat immature Leydig cells.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Androgens/biosynthesis , Leukemia Inhibitory Factor/pharmacology , Leydig Cells/metabolism , Phosphoproteins/metabolism , 17-Hydroxysteroid Dehydrogenases/genetics , Animals , Cells, Cultured , Leydig Cells/cytology , Leydig Cells/drug effects , Luteinizing Hormone/pharmacology , Male , Phosphoproteins/genetics , Rats , Up-Regulation
12.
Neurodegener Dis ; 16(5-6): 357-69, 2016.
Article in English | MEDLINE | ID: mdl-27228974

ABSTRACT

BACKGROUND: Basic fibroblast growth factor (bFGF) has been increasingly investigated due to its neuroprotection in neurodegenerative disorders. Because there are still no cures for any of these disorders, it is crucial to identify new therapeutic targets and screen potential drugs. The increased phosphorylation of tau at Ser396 leads to intracellular tau accumulation, which forms neurofibrillary tangles in Parkinson's disease (PD). In this study, neuroprotection by bFGF was observed, and the mechanisms related to its regulation of phosphorylated tau were investigated. METHODS: bFGF-loaded liposome carriers were intranasally administered to rats. The neuroprotective effects of bFGF were assessed in a PD model induced by 6-hydroxydopamine (6-OHDA) in vivo and in vitro. The phosphorylation of tau was measured, and the PI3K/Akt-GSK3ß signaling pathway was investigated. RESULTS: Our study demonstrated that liposomes markedly assisted in the delivery of bFGF to the striatum and substantia nigra of rats and enhanced the neuroprotective effects of bFGF on dopaminergic neurons. bFGF treatment significantly ameliorated the behavioral deficits induced by 6-OHDA, rescued the loss of tyrosine hydroxylase-positive neurons and increased the number of Nissl bodies. bFGF reduced the phosphorylation of tau and GSK3ß and increased the phosphorylation of PI3K/Akt. CONCLUSION: Liposomes markedly assisted in the delivery of bFGF to the brain and enhanced the neuroprotective effects of bFGF by inhibiting the phosphorylation of tau. bFGF down-regulated the phosphorylation of tau by increasing the phosphorylation of GSK3ß via the PI3K/Akt signaling pathway. These findings provide a new vision of bFGF as a potential therapy for PD.


Subject(s)
Brain/metabolism , Fibroblast Growth Factor 2/administration & dosage , Neuroprotective Agents/administration & dosage , Parkinson Disease/metabolism , Signal Transduction/drug effects , tau Proteins/metabolism , Administration, Intranasal , Animals , Brain/drug effects , Cell Line, Tumor , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Drug Carriers/administration & dosage , Drug Carriers/pharmacology , Fibroblast Growth Factor 2/therapeutic use , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Liposomes/administration & dosage , Liposomes/pharmacology , Male , Motor Activity/drug effects , Neuroprotective Agents/therapeutic use , Oxidopamine , Parkinson Disease/drug therapy , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism
13.
Biol Reprod ; 92(3): 77, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25609837

ABSTRACT

Cyclin-dependent kinase inhibitors p21(Cip1) (CDKN1A) and p27(Kip1) (CDKN1B) are expressed in Leydig cells. Previously, we reported that Cdkn1b knockout in the mouse led to increased Leydig cell proliferative capacity and lower steroidogenesis. However, the relative importance of CDKN1A and CDKN1B in these regulations was unclear. In the present study, we examined the relative importance of CDKN1A and CDKN1B in regulation of Leydig cell proliferation and steroidogenesis by whole-body knockout of CDKN1A (Cdkn1a(-/-)) and CDKN1A/CDKN1B double knockout (DBKO). The cell number, 5-bromo-2-deoxyuridine incorporation rate, steroidogenesis, and steroidogenic enzyme mRNA levels and activities of Leydig cells were compared among wild-type (WT), Cdkn1a(-/-), and DBKO mice. Relative to WT mice, Leydig cell number per testis was doubled in the DBKO and unchanged in the Cdkn1a(-/-) mice. Testicular testosterone levels and mRNA levels for luteinizing hormone receptor (Lhcgr), steroidogenic acute regulatory protein (Star), cholesterol side-chain cleavage enzyme (Cyp11a1), 17alpha-hydroxylase/17,20-lyase (Cyp17a1), and 17beta-hydroxysteroid dehydrogenase 3 (Hsd17b3) and their respective proteins were significantly lower in the DBKO mice. However, testicular testosterone level was unchanged in the Cdkn1a(-/-) mice, although Lhcgr mRNA levels were significantly lower relative to those in the WT control. We conclude that Cdkn1a(-/-) did not increase Leydig cell numbers (although a defect of Leydig cell function was noted), whereas DBKO caused a significant increase of Leydig cell numbers but a decrease of steroidogenesis.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Leydig Cells/cytology , Sexual Maturation/physiology , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/physiology , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/physiology , Leydig Cells/metabolism , Male , Mice , Mice, Knockout , Models, Animal , Phosphoproteins/metabolism , RNA, Messenger/metabolism , Receptors, LH/metabolism , Sexual Maturation/genetics , Steroids/metabolism
14.
Appl Microbiol Biotechnol ; 99(14): 5997-6007, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25758955

ABSTRACT

Nogo-66, a hydrophilic loop of 66 amino acids flank two hydrophobic domains of the Nogo-A C terminus, interacts with the Nogo-66 receptor (NgR) to exert numerous functions in the central nervous system (CNS). Nogo-66 has important roles in aspects of neuronal development, including cell migration, axon guidance, fasciculation, and dendritic branching, and in aspects of CNS plasticity, including oligodendrocyte differentiation and myelination. Here, the small ubiquitin-related modifier (SUMO) was fused to the target gene, Nogo-66, and the construct was expressed in Escherichia coli (E. coli). Under the optimal fermentation conditions, the soluble expression level of the fusion protein was 33 % of the total supernatant protein. After cleaving the fusion proteins with SUMO protease and purifying them by Ni-NTA affinity chromatography, the yield and purity of recombinant Nogo-66 obtained by 10-L scale fermentation were 23 ± 1.5 mg/L and greater than 93 %, respectively. The authenticity of the recombinant Nogo-66 was confirmed by an electrospray ionization-mass spectrometry analysis. The functional analyses indicated that the recombinant Nogo-66 was capable of binding the NgR specifically. The immunofluorescence results showed that the recombinant Nogo-66 could significantly inhibit neurite outgrowth of rat pheochromocytoma (PC12) cells stimulated by nerve growth factor and cerebellar granule cells (CGCs). Furthermore, Nogo-66 inhibited neurite outgrowth by increasing the level of phosphorylated Rho-associated coiled-coil-containing protein kinase 2 (ROCK2), collapsin response mediator protein 2 (CRMP2), and myosin light chain (MLC). This study provided a feasible and convenient production method for generating sufficient recombinant Nogo-66 for experimental and clinical applications.


Subject(s)
Myelin Proteins/metabolism , Neurites/drug effects , SUMO-1 Protein/metabolism , Animals , Cell Line, Tumor , Chromatography, Affinity , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Intercellular Signaling Peptides and Proteins , Myelin Proteins/genetics , Myelin Proteins/isolation & purification , Myosin Light Chains/metabolism , Nerve Tissue Proteins/metabolism , Nogo Proteins , Protein Binding , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , SUMO-1 Protein/genetics , Spectrometry, Mass, Electrospray Ionization , rho-Associated Kinases/metabolism
15.
Biochem Biophys Res Commun ; 445(4): 795-801, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24530908

ABSTRACT

It has been reported that acidic fibroblast growth factor (aFGF) is expressed in breast cancer and via interactions with fibroblast growth factor receptors (FGFRs) to promote the stage and grade of the disease. Thus, aFGF/FGFRs have been considered essential targets in breast cancer therapy. We identified a specific aFGF-binding peptide (AGNWTPI, named AP8) from a phage display heptapeptide library with aFGF after four rounds of biopanning. The peptide AP8 contained two (TP) amino acids identical and showed high homology to the peptides of the 182-188 (GTPNPTL) site of high-affinity aFGF receptor FGFR1. Functional analyses indicated that AP8 specifically competed with the corresponding phage clone A8 for binding to aFGF. In addition, AP8 could inhibit aFGF-stimulated cell proliferation, arrested the cell cycle at the G0/G1 phase by increasing PA2G4 and suppressing Cyclin D1 and PCNA, and blocked the aFGF-induced activation of Erk1/2 and Akt kinase in both breast cancer cells and vascular endothelial cells. Therefore, these results indicate that peptide AP8, acting as an aFGF antagonist, is a promising therapeutic agent for the treatment of breast cancer.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Fibroblast Growth Factor 1/metabolism , Peptides/chemistry , Peptides/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Breast/drug effects , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Female , Humans , MAP Kinase Signaling System/drug effects , Peptide Library , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA-Binding Proteins/metabolism
16.
Appl Microbiol Biotechnol ; 98(18): 7837-44, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24728756

ABSTRACT

Pramlintide is an artificially designed protein which has the same function as amylin in human body. This protein is extremely difficult to synthesize through prokaryotic expression method because of its two essential active sites, intrachain disulfide bond and C-terminal amide group. Since α-amidating monooxygenase is widely distributed in human and animal, it is possible to use pramlintide precursor with an additional C-terminal glycine (PAG), which is the potential substrate of α-amidating monooxygenase, for in vivo applications. The recombinant PAG was expressed in Escherichia coli using the small ubiquitin-related modifier (SUMO) as the molecular chaperone, and the optimal fusion expression level reached to 36.3% of the total supernatant protein. Under optimal conditions in a 10-L fermentor, the recombinant PAG was obtained with a purity of greater than 95%, and the average expression level was reached to 20 mg/L. The authenticity and the intrachain disulfide bridge of PAG were confirmed by Western blotting and matrix-assisted laser desorption/ionization coupled to time-of-flight mass spectrometry (MALDI-TOF MS) as well as N-terminal sequencing of protein. Based on an L6 myoblast cell model in vitro and an animal model of gastric emptying in vivo, the results of activity revealed that PAG showed a lower biological activity in vitro but has almost the same activity as the chemically synthesized pramlintide in vivo.


Subject(s)
Escherichia coli/metabolism , Islet Amyloid Polypeptide/metabolism , Islet Amyloid Polypeptide/pharmacology , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Animals , Cell Line , Gastric Emptying/drug effects , Islet Amyloid Polypeptide/genetics , Male , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics
17.
Biol Pharm Bull ; 37(7): 1149-57, 2014.
Article in English | MEDLINE | ID: mdl-24989006

ABSTRACT

Disabilities triggered by neurodegeneration mainly result in mortality in the elderly, and patients with neurodegenerative disease also display deficits in olfactory function. Therefore drug distribution to the brain through intranasal administration has become one of the most difficult challenges in the treatment of central nervous system (CNS) diseases. TAT-human acidic fibroblast growth factor (HaFGF) is a new fused protein retaining the neuroprotective activities of HaFGF, and is a promising prospect in the treatment of neurodegenerative diseases. TAT (a cell-penetrating peptide) contains a high relative abundance of positively charged amino acids such as lysine and arginine, which have a powerful attraction to the negatively charge on the nasal epithelial membrane. The present study focused on the evaluation of the safety and absorption characteristics of TAT-HaFGF following intranasal administration. After TAT-HaFGF intranasal administration (100, 300, 600 µg/kg) for 5 weeks, hematoxylin-eosin (HE) staining showed no pathology in any of the investigated tissues and organs. The expression of olfactory marker protein (OMP) was observed with immunohistochemical staining, which showed no altered expression in the sensory neurons of the nasal epithelium. Nasal ciliotoxicity studies carried out using an in situ palate model and optical microscope showed that TAT-HaFGF had no nasal ciliotoxicity. The distribution of the TAT-HaFGF following intranasal administration was assessed using a radioisotopic tracing method. Radioactivity was observed in the brain after 15 min. This became stronger at 30 min and weaker at 1 h. All of the results confirmed the in vivo safety of TAT-HaFGF via intranasal administration.


Subject(s)
Brain/drug effects , Fibroblast Growth Factors/administration & dosage , Gene Products, tat/administration & dosage , Nasal Absorption/drug effects , Nasal Mucosa/drug effects , Neuroprotective Agents/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Administration, Intranasal , Animals , Brain/metabolism , Bufo bufo , Cilia/drug effects , Female , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/pharmacokinetics , Gene Products, tat/adverse effects , Gene Products, tat/pharmacokinetics , Male , Nasal Mucosa/metabolism , Neuroprotective Agents/adverse effects , Neuroprotective Agents/pharmacokinetics , Olfactory Bulb/drug effects , Olfactory Bulb/metabolism , Rats, Sprague-Dawley , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/pharmacokinetics , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Tissue Distribution
18.
Cell Biochem Biophys ; 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39342069

ABSTRACT

Periodontitis is a prevalent condition characterized by inflammation and tissue destruction within the periodontium, with hypoxia emerging as a contributing factor to its pathogenesis. Hypoxia-inducible factor 1α (HIF-1α) has a crucial role in orchestrating adaptive responses to hypoxic microenvironments and has been implicated in various inflammatory-related diseases. Understanding the interplay between HIF-1α, matrix metalloproteinases (MMPs), and inflammatory responses in periodontitis could provide insights into its molecular mechanisms. We investigated the relationship between HIF-1α, MMP2, and MMP9 in gingival crevicular fluid (GCF) and periodontal ligament stem cells (PDLSCs) from periodontitis patients. The expression levels of HIF-1α, MMP2, MMP9, and inflammatory factors (IL-6, IL-1ß, TNF-α) were assessed using enzyme-linked immunosorbent assay (ELISA) and real-time PCR (RT-PCR). Additionally, osteogenic differentiation of PDLSCs was identified by alkaline phosphatase activity. Significantly elevated levels of HIF-1α, MMP2, and MMP9 were observed in GCF of periodontitis patients compared to controls. Positive correlations were found between HIF-1α and MMP2/MMP9, as well as with IL-6, IL-1ß, and TNF-α. Modulation of HIF-1α expression in PDLSCs revealed its involvement in MMP2/9 secretion and inflammatory responses, with inhibition of HIF-1α mitigating these effects. Furthermore, HIF-1α inhibition alleviated the reduction in osteogenic differentiation induced by inflammatory stimuli. Our findings elucidate the regulatory role of HIF-1α in MMP expression, inflammatory responses, and osteogenic differentiation in periodontitis. In conclusion, targeting HIF-1α signaling pathways may offer therapeutic opportunities for managing periodontitis and promoting periodontal tissue regeneration.

19.
Bioengineering (Basel) ; 11(6)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38927810

ABSTRACT

Excessive dietary fat intake is closely associated with an increased risk of obesity, type 2 diabetes, cardiovascular disease, gastrointestinal diseases, and certain types of cancer. The administration of multi-strain probiotics has shown a significantly beneficial effect on the mitigation of obesity induced by high-fat diets (HFDs). In this study, Amuc_1100, an outer membrane protein of Akkermansia muciniphila, was fused with green fluorescent protein and LPXTG motif anchor protein and displayed on the surface of Lactobacillus rhamnosus (pLR-GAA) and Lactobacillus plantarum (pLP-GAA), respectively. The localization of the fusion protein on the bacterial cell surface was confirmed via fluorescence microscopy and Western blotting. Both recombinant strains demonstrated the capacity to ameliorate hyperglycemia and decrease body weight gain in a dose-dependent manner. Moreover, daily oral supplementation of pLR-GAA or pLP-GAA suppressed the HFD-induced intestinal permeability by regulating the mRNA expressions of tight junction proteins and inflammatory cytokines, thereby reducing gut microbiota-derived lipopolysaccharide concentration in serum and mitigating damage to the gut, liver, and adipose tissue. Compared with Lactobacillus rhamnosus treatment, high-dose pLR-GAA restored the expression level of anti-inflammatory factor interleukin-10 in the intestine. In conclusion, our approach enables the maintenance of intestinal health through the use of recombinant probiotics with surface-displayed functional protein, providing a potential therapeutic strategy for HFD-induced obesity and associated metabolic comorbidities.

20.
Biochem Biophys Res Commun ; 436(2): 300-5, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23743199

ABSTRACT

Nerve growth factor (NGF) has been reported to be involved in male reproductive physiology. However, few reports have described the activity of NGF during Leydig cell development. The objective of the present study was to examine the role of NGF during stem-Leydig-cell (SLC) regeneration. We investigated the effects of NGF on Leydig-cell (LC) regeneration by measuring mRNA levels in the adult rat testis after ethane dimethanesulfonate (EDS) treatment. Furthermore, we used the established organ culture model of rat seminiferous tubules to examine the regulation of NGF during SLC proliferation and differentiation using EdU staining, real-time PCR and western blotting. Progenitor Leydig cells (PLCs) and immature Leydig cells (ILCs) were also used to investigate the effects of NGF on LCs at different developmental stages. NGF mRNA levels changed significantly during Leydig-cell regeneration in vivo. In vitro, NGF significantly promoted the proliferation of stem Leydig cells and also induced steroidogenic enzyme gene expression and 3ß-HSD protein expression. The data from PLCs and ILCs showed that NGF could increase Cyclin D1 and Hsd 17b3 mRNA levels in PLCs and Cyclin D1 mRNA levels in ILCs. These results indicate that NGF may play an important role during LC regeneration by regulating the proliferation and differentiation of LCs at different developmental stages, from SLCs to PLCs and from PLCs to ILCs. The discovery of this effect of NGF on Leydig cells will provide useful information for developing new potential therapies for PADAM (Partial Androgen Deficiency in the Aging Male).


Subject(s)
Cell Differentiation/genetics , Cell Proliferation , Leydig Cells/metabolism , Nerve Growth Factor/genetics , Regeneration/genetics , Stem Cells/metabolism , Animals , Blotting, Western , Cell Differentiation/drug effects , Cells, Cultured , Cyclin D1/genetics , Cyclin D1/metabolism , Gene Expression/drug effects , Leydig Cells/cytology , Leydig Cells/drug effects , Male , Mesylates/pharmacology , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Organ Culture Techniques , Phosphoproteins/genetics , Phosphoproteins/metabolism , Progesterone Reductase/genetics , Progesterone Reductase/metabolism , Rats , Rats, Sprague-Dawley , Regeneration/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Seminiferous Tubules/cytology , Seminiferous Tubules/drug effects , Seminiferous Tubules/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Steroid Isomerases/genetics , Steroid Isomerases/metabolism , Testolactone/analogs & derivatives , Testolactone/blood , Testolactone/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL